WO2006133242A2 - Azinone and diazinone v3 inhibitors for depression and stress disorders - Google Patents

Azinone and diazinone v3 inhibitors for depression and stress disorders Download PDF

Info

Publication number
WO2006133242A2
WO2006133242A2 PCT/US2006/022025 US2006022025W WO2006133242A2 WO 2006133242 A2 WO2006133242 A2 WO 2006133242A2 US 2006022025 W US2006022025 W US 2006022025W WO 2006133242 A2 WO2006133242 A2 WO 2006133242A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound according
chosen
cycloalkyl
optionally substituted
Prior art date
Application number
PCT/US2006/022025
Other languages
French (fr)
Other versions
WO2006133242A3 (en
Inventor
Jeffrey John Letourneau
Koc-Kan Ho
Michael J. Ohlmeyer
Patrick Jokiel
Christopher Mark Riviello
Original Assignee
Pharmacopeia, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pharmacopeia, Inc. filed Critical Pharmacopeia, Inc.
Priority to AU2006255009A priority Critical patent/AU2006255009B2/en
Priority to CA002610400A priority patent/CA2610400A1/en
Priority to MX2007015427A priority patent/MX2007015427A/en
Priority to EP06760716.8A priority patent/EP1890697B1/en
Priority to BRPI0611348-6A priority patent/BRPI0611348A2/en
Priority to CN2006800237203A priority patent/CN101212969B/en
Priority to JP2008515847A priority patent/JP5002592B2/en
Publication of WO2006133242A2 publication Critical patent/WO2006133242A2/en
Publication of WO2006133242A3 publication Critical patent/WO2006133242A3/en
Priority to IL187634A priority patent/IL187634A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/46Drugs for disorders of the endocrine system of the suprarenal hormones for decreasing, blocking or antagonising the activity of glucocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/26Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/34One oxygen atom
    • C07D239/36One oxygen atom as doubly bound oxygen atom or as unsubstituted hydroxy radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/08Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms
    • C07D295/084Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/088Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention relates to a chemical class of substituted pyridines, pyrimidines, pyrazines, pyridinones, pyrimidinones, pyrazinones and phenylacetamides useful in treating depression, stress and other disorders.
  • hypothalamo-pituitary-adrenal (HPA) axis is the major stress axis in humans and other mammals.
  • a variety of stressors and multiple other classes of stimuli) cause release of the hormone ACTH (adrenocorticotropic hormone) from the anterior pituitary gland.
  • ACTH enters the systemic circulation and acts on the adrenal cortex to promote synthesis and release of glucocorticoid hormone (the major endogenous glucocorticoid being Cortisol in humans and corticosterone in rodents).
  • the glucocorticoids exert a broad spectrum of effects, the main purpose of which is to mobilize energy sources for successful responsiveness and eventual adaptation to the stressor.
  • dysregulation of the HPA axis is a reflection of enhanced vulnerability and poor adaptation to chronic stress and that chronic stress therefore plays a major role in the development of affective illness (Sperry and Carlson, DSM-IV diagnosis to treatment, 2 nd Edition, Taylor & Francis, 1996).
  • This central concept is supported by experimental evidence utilizing animal models of chronic stress, where aberrant HPA function closely resembles that seen in clinical settings (De Goeij et al, Neuroendocrinology, 1991, 53, 150-159; Plotsky and Meaney, MoI. Brain Res. 1993, 18, 195-200).
  • CRH corticotropin releasing hormone
  • AVP arginine vasopressin
  • the HPA axis is most potently activated by psychological stressors ⁇ i.e., those which require a cognitive assessment of the stimulus).
  • the patterns of AVP and CRH release vary as a function of the type of stressor involved. Acute stress, whether physical or psychological, elicits rapid and robust CRH release. For several psychological stressors, however, chronic application elicits enhanced AVP storage in the median eminence, increased niRNA synthesis, and reduction in AVP neurosecretory granules, whereas similar markers of CRH synthesis and release are relatively unaffected.
  • V3 antagonists may be useful in the treatment of aggressive behavior [see Wersinger et al. MoI. Psychiatry 7. 975- 984 (2002); Blanchard et al. Pharmcol. Biochem. Behav. 80. 189-194 (2005); and Wersinger et al. Horm. Behav. 46, 638-645 (2004)]; insomnia in elderly patients [see Kalamatianos et al. J. Neuroendocrinol. 16, 493-501 (2004)]; cancer [see Dahia et al. J. Clin. Endocrin. Metab. 81. 1768-1771 (1996)]; Cushing's Disease [see Perraudin et al. J. Clin. Endocrin.
  • vasopressin In addition to the V3 receptor, vasopressin also activates peripheral receptors, i.e., the Via receptor, predominantly found on liver and vascular tissue and the V2 receptor, predominantly found on kidney tissue. Interaction at these receptors mediates the pressor and antidiuretic actions of AVP.
  • peripheral receptors i.e., the Via receptor
  • the Via receptor predominantly found on liver and vascular tissue
  • the V2 receptor predominantly found on kidney tissue. Interaction at these receptors mediates the pressor and antidiuretic actions of AVP.
  • X 4 -X 5 is chosen from CR 4 -CR 5 , N-CR 5 and CR 4 -N; in genus in, one of X 6 , X 7 and X 8 is N and the other two are CR 4 and CR 5 .
  • R 1 is chosen from (C 1- io)alkyl, (Cs. ⁇ cycloalkyl, [(C 3-1 o)cycloalkyl(Ci -2 )alkyl], said (C 1-1 o)alkyl, (C 3-1 o)cycloalkyl, and [(C 3-10 )cycloalkyl (C 1-2 )alkyl] being optionally substituted with one or more halogens, (Q ⁇ alkoxy, (C 2- 6 )alkenyl, (C 2-6 )alkynyl, phenyl or benzyl; Ar is chosen from (C 1- io)alkyl, (Cs. ⁇ cycloalkyl, [(C 3-1 o)cycloalkyl(Ci -2 )alkyl], said (C 1-1 o)alkyl, (C 3-1 o)cycloalkyl, and [(C 3-10 )cycloalkyl (C 1-2 )alkyl] being optionally
  • R 4 and R 5 are independently chosen from H, (Ci -6 )alkyl, (Ci ⁇ alkyloxy or halogen, said (Ci ⁇ alkyl and (Ci ⁇ alkyloxy being optionally substituted with one or more halogens;
  • G is a linking moiety spanning 4 to 7 atoms between termini; and R 8 and R 9 are residues that, in combination, maintain the basicity of N.
  • R 1 When X ⁇ -X 5 is CR 4 -N and G is alkylene, R 1 must be chosen from (Ci -6 )alkyl, (C 3-6 )cycloalkyl and [(C 3-6 )cycloalkyl(Ci -2 )alkyl].
  • R 1 is not restricted to this subset of values, G is a linking moiety incorporating at least one of (a) an sp2 hybridized carbon; or (b) a cyclic structure.
  • G may additionally be -N(R 10 )-(C 4-6 )alkylene for all values of R 1 .
  • R 10 is H or (C 1-6 )alkyl.
  • a subgenus of compounds in accordance with embodiments of the invention is the genus I:
  • the genus I can be divided into three subgenera:
  • compositions comprising a pharmaceutically acceptable carrier and a compound as described above.
  • pyridines pyrimidines, pyrazines, pyridinones, pyrimidinones, pyrazinones and phenylacetamides falling within a general formula
  • B represents a six-membered, planar carbocycle or planar nitrogen heterocycle.
  • G is a linking moiety spanning 4 to 7 atoms between termini.
  • the -NR 8 R 9 must be 4 to 7 atoms removed from the ring B.
  • the precise constituents of G are not critical.
  • G will incorporate either an sp2 hybridized carbon or a cyclic structure.
  • R 8 and R 9 together form a 4- to 7-menibered nitrogenous heterocycle, G may be -N(R 10 )-(C 4 . 6 )alkylene.
  • G may also be an optionally substituted carbocycle or heterocycle, attached to the B ring and to -NR 8 R 9 by a direct bond or by a C 1 -C 5 alkylene chain.
  • G may also be an optionally substituted nitrogenous heterocycle, attached to the B ring by a direct bond or by a C 1 -C 5 alkylene chain; in this case a nitrogen of the nitrogenous heterocycle may correspond to -NR 8 R 9 so that R 9 becomes formally part of G.
  • R and R may also be taken together and attach to G so as to form a nitrogen-containing heterocycle, e.g. a pyridine ring attached at one of the ring carbon atoms to the linker which links the pyridine ring to ring B. In such cases, there will be 4 to 7 atoms between the ring B and the carbon atom of the nitrogen-containing heterocycle.
  • the residues R 8 and R 9 must maintain the basicity of N.
  • alkyl residues of various sorts are within the invention; alkylene and similar residues (e.g. alkylene with heteroatom interruption) that tie the nitrogen into a ring are within the invention. Even residues that introduce aromaticity are tolerated, as long as the nitrogen remains basic (e.g. pyridine).
  • R 8 and R 9 may be hydrogen.
  • a genus in accordance with some embodiments of the invention comprises pyridinones, pyrimidinones and pyrazinones that fall within the general formula I above:
  • X 5 is CR 4 -CR 5 , N-CR 5 or CR 4 -N.
  • subgenera in accordance with embodiments of the invention include the subgenus in which -NR 8 R 9 is a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings, preferably a piperidine or morpholine, and G is a (Cs. ⁇ hydrocarbon chain attached to the pyridinone, pyrimidinone or pyrazinone through an amide or amine:
  • E is (C 2-1 o)hydrocarbon; and — is a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings.
  • the nitrogenous heterocycle may be substituted, for example, 4-hydroxypiperidin-l-yl, 4-hydroxy-4- methylpiperidin-1-yl and 4,4-dimethylpiperidin-l-yl.
  • the (C 2- io)hydrocarbon may be straight chain, branched or cyclic as long as the 4-7 atom spacing between -NR 8 R 9 and the pyridinone, pyrimidinone or pyrazinone ring is maintained. Examples of species in this subgenus include:
  • a further example of a subgenus in accordance with embodiments of the invention is the subgenus in which -NR 8 R 9 is attached via a direct bond or (Ci- 6 )hydrocarbon, J, to a single ring carbocycle or heterocycle of 4 to 7 atoms or a
  • the carbocycle or heterocycle is directly attached to the pyridinone, pyrimidinone or pyrazinone:
  • nitrogenous heterocycle e.g. oxadiazolyl, pyrrolidinyl and piperidinyl
  • J is methylene, ethylene or propylene
  • -NR 8 R 9 is chosen from piperidine, morpholine and -N[C 1-3 alkyl] 2 .
  • species in this subgenus include:
  • a further example of a subgenus in accordance with embodiments of the invention is the subgenus in which R is alkylene or similar residue (e.g. alkylene with heteroatom interruption) "tied back" into a nitrogen that is directly attached to the pyridinone, pyrimidinone or pyrazinone ring:
  • hexahydro-l,4-diazepine ring is a hexahydro-l,4-diazepine ring.
  • An example is
  • a further example of a subgenus in accordance with embodiments of the invention is the subgenus in which R 9 is alkylene or similar residue (e.g. alkylene with heteroatom interruption) "tied back" into a chain that is attached to the pyridinone, pyrimidinone or pyrazinone ring through an amine or amide linkage.
  • R 9 is alkylene or similar residue (e.g. alkylene with heteroatom interruption) "tied back" into a chain that is attached to the pyridinone, pyrimidinone or pyrazinone ring through an amine or amide linkage.
  • R 8 is C 1-10 hydrocarbon.
  • This particular subgenus also includes compounds of the formulae in which R 1 is C 3 -C 6 alkyl.
  • R 1 is C 3 -C 6 alkyl.
  • R 8 and R may be taken together and attach to G so as to form a nitrogen-containing heterocycle.
  • Pyr represents imidazole, pyridine attached through a carbon, substituted imidazole or substituted pyridine attached through a carbon.
  • An example is:
  • R 1 may be chosen from C 3-6 alkyl and cycloalkyl and Ci -3 alkyl substituted with phenyl, methoxy or alkynyl.
  • R 1 may be t-butyl, isopropyl, cyclopentyl, ⁇ -methylbenzyl, methoxypropyl or propargyl.
  • Ar may be chosen from phenyl and phenyl substituted with halogen (e.g. chloro and fluoro), Ci -2 alkyl, (e.g. methyl) trifluoromethyl, C 1-3 alkyloxy (e.g. methoxy), C 1-4 cycloalkyloxy or trifluoromethoxy.
  • Ar is a 3-substituted phenyl ring, for example a substituted phenyl ring selected from 3-chlorophenyl, 3- fiuorophenyl, 3-methoxyphenyl, 3-trifluoromethoxyphenyl, 3-chloro-4- fluorophenyl, 4-fluoro-3-methoxyphenyl and 3,5-dimethoxyphenyl.
  • the compounds in accordance with embodiments of the invention all contain a basic nitrogen, they may be presented as salts.
  • reference to the compound includes its salts.
  • pharmaceutically acceptable salt refers to salts whose counter ion derives from pharmaceutically acceptable non-toxic acids and bases.
  • Suitable pharmaceutically acceptable base addition salts for the compounds of the present invention include inorganic acids, and organic acids.
  • Examples include acetate, benzenesulfonate (besylate), benzoate, bicarbonate, bisulfate, carbonate, camphorsulfonate, citrate, ethanesulfonate, fumarate, gluconate, glutamate, glycolate, bromide, chloride, isethionate, lactate, maleate, malate, mandelate, methanesulfonate, mucate, nitrate, pamoate, pantothenate, phosphate, succinate, sulfate, tartrate, trifluoroacetate, p-toluenesulfonate, acetamidobenzoate, adipate, alginate, aminosalicylate, anhydromethylenecitrate, ascorbate, aspartate, calcium edetate, camphorate, camsylate, caprate, caproate, caprylate, cinnamate, cyclamate, dichloroacetate, ede
  • suitable pharmaceutically acceptable base addition salts for the compounds of the present invention include ammonium, metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, N 5 N'- dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine.
  • base addition salts includes those made from: arecoline, arginine, barium, benethamine, benzathine, betaine, bismuth, clemizole, copper, deanol, diethylamine, diethylaminoethanol, epolamine, ethylenediamine, ferric, ferrous, glucamine, glucosamine, histidine, hydrabamine, imidazole, isopropylamine, manganic, manganous, methylglucamine, morpholine, morpholineethanol, n- ethylmorpholine, n-ethylpiperidine, piperazine, piperidine, polyamine resins, purines, theobromine, triethylamine, trimethylamine, tripropylamine, trolamine, and tromethamine.
  • Alkyl is intended to include linear, branched, or cyclic hydrocarbon structures and combinations thereof. When not otherwise restricted, the term refers to alkyl of 20 or fewer carbons. Lower alkyl refers to alkyl groups of 1-6 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s-and t-butyl and the like. Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of 3-8 carbon atoms. Examples of cycloalkyl groups include c-propyl, c-butyl, c-pentyl, norbornyl, adamantyl and the like. In accordance with standard nomenclature, the term "alkylene” applies to alkyl residues having two points of attachment. For example, propylene refers to -CH 2 CH 2 CH 2 -.
  • hydrocarbon includes alkyl, cycloalkyl, alkenyl, alkynyl, aryl and combinations thereof. Examples include benzyl, phenethyl, cyclohexylmethyl, camphoryl and naphthylethyl.
  • Alkoxy or alkoxyl refers to groups of 1-8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to four carbons.
  • Oxaalkyl refers to alkyl residues in which one or more carbons (and their associated hydrogens) have been replaced by oxygen. Examples include methoxypropoxy, 3,6,9-trioxadecyl and the like.
  • the term oxaalkyl is intended as it is understood in the art [see Naming and Indexing of Chemical Substances for Chemical Abstracts, published by the American Chemical Society, ⁇ fI 96, but without the restriction of Tfl27(a)], i.e. it refers to compounds in which the oxygen is bonded via a single bond to its adjacent atoms (forming ether bonds).
  • thiaalkyl and azaalkyl refer to alkyl residues in which one or more carbons have been replaced by sulfur or nitrogen, respectively. Examples include ethylaminoethyl and methylthiopropyl.
  • Acyl refers to groups of 1-8 carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality.
  • One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl. Examples include formyl, acetyl, propionyl, isobutyryl, ⁇ -butoxycarbonyl, benzoyl, benzyloxycarbonyl and the like.
  • Lower-acyl refers to groups containing one to four carbons.
  • Aryl and heteroaryl refer to aromatic or heteroaromatic rings, respectively, as substituents.
  • Heteroaryl contains one, two or three heteroatoms selected from O, N, or S. Both refer to monocyclic 5- or 6-membered aromatic or heteroaromatic rings, bicyclic 9- or 10-membered aromatic or heteroaromatic rings and tricyclic 13- or 14-membered aromatic or heteroaromatic rings.
  • Aromatic 6-14-membered carbocyclic rings include, e.g., benzene, naphthalene, indane, tetralin, and fluorene and the 5-10-membered aromatic heterocyclic rings include, e.g., imidazole, pyridine, indole, thiophene, benzopyranone, thiazole, furan, benzimidazole, quinoline, isoquinoline, quinoxaline, pyrimidine, pyrazine, tetrazole and pyrazole.
  • Arylalkyl means an alkyl residue attached to an aryl ring. Examples are benzyl, phenethyl and the like.
  • Substituted alkyl, aryl, cycloalkyl, heterocyclyl etc. refer to alkyl, aryl, cycloalkyl, or heterocyclyl wherein up to three H atoms in each residue are replaced with halogen, haloalkyl, hydroxy, loweralkoxy, carboxy, carboalkoxy (also referred to as alkoxycarbonyl), carboxamido (also referred to as alkylaminocarbonyl), cyano, carbonyl, nitro, amino, alkylamino, dialkylamino, mercapto, alkylthio, sulfoxide, sulfone, acylamino, amidino, phenyl, benzyl, heteroaryl, phenoxy, benzyloxy, or heteroaryloxy.
  • halogen means fluorine, chlorine, bromine or iodine.
  • R-groups may form rings or heterocycles.
  • R 8 and R 9 together form a 4- to 7-membered nitrogenous heterocycle. It is intended that these rings may exhibit various degrees of unsaturation, may include heteroatoms and may be substituted with lower alkyl or alkoxy.
  • the compounds of this invention can exist in radiolabeled form, i.e., the compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • Radioisotopes of hydrogen, carbon, phosphorous, fluorine, and chlorine include 3 H, 14 C, 35 S, 18 F, and 36 Cl, respectively.
  • Compounds that contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of embodiments of the invention. Tritiated, i.e. 3 H, and carbon-14, i.e., 14 C, radioisotopes are particularly known for their ease in preparation and detectability.
  • Radiolabeled compounds of this invention can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabeled compounds can be prepared by carrying out the procedures disclosed in the Examples and Schemes by substituting a readily available radiolabeled reagent for a non-radiolabeled reagent. Radiolabeled compounds are useful in screens for V3 agonists and antagonists.
  • the compounds described herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms.
  • Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. Included in embodiments of the present invention are all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
  • Me, Et, Ph, Tf, Ts and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, toluenesulfonyl and methanesulfonyl respectively.
  • the following abbreviations and terms have the indicated meanings throughout:
  • EDC, EDCI 1 -ethyl-3 -(3 '-dimethylaminopropyl)carbodiimide
  • NMO N-methylmorpholine oxide
  • Ph phenyl
  • PPTS pyridinium p-toluenesulfonate
  • a pharmaceutical composition comprising a compound as described herein or a pharmaceutically acceptable salt or solvate thereof, together with one or more pharmaceutically carriers thereof and optionally one or more other therapeutic ingredients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular), rectal and topical (including dermal, buccal, sublingual and intraocular) administration.
  • the most suitable route may depend upon the condition and disorder of the recipient.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods of treatment in accordance with embodiments of the invention include the step of bringing into association a compound in accordance with embodiments of the invention or a pharmaceutically acceptable salt or solvate thereof ("active ingredient”) with the carrier, which constitutes one or more accessory ingredients.
  • active ingredient a compound in accordance with embodiments of the invention or a pharmaceutically acceptable salt or solvate thereof
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations in accordance with embodiments of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in- water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein.
  • the pharmaceutical compositions may include a "pharmaceutically acceptable inert carrier", and this expression is intended to include one or more inert excipients, which include starches, polyols, granulating agents, macrocrystalline cellulose, diluents, lubricants, binders, disintegrating agents, and the like. If desired, tablet dosages of the disclosed compositions may be coated by standard aqueous or nonaqueous techniques, "Pharmaceutically acceptable carrier” also encompasses controlled release means.
  • compositions in accordance with embodiments of the present invention may also optionally include other therapeutic ingredients, anti-caking agents, preservatives, sweetening agents, colorants, flavors, desiccants, plasticizers, dyes, and the like. Any such optional ingredient must, of course, be compatible with the compound of the invention to insure the stability of the formulation.
  • the dose range for adult humans is generally from 0.005 mg to 10 g/day orally. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of a compound or mixture of compounds in accordance with embodiments of the invention which is or are effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the precise amount of compound or compounds administered to a patient will be the responsibility of the attendant physician. However, the dose employed will depend on a number of factors, including the age and sex of the patient, the precise disorder being treated, and its severity.
  • Carboxylic acid 3-2 was prepared from alkene 3-1 (159 mg; 0.378 mmol) using the general procedures analogous to those described in Example 2, Steps 3 and 4. This afforded 130 mg (78% for 2 steps from 3-1) of acid 3-2.
  • Compound 7-1 was prepared from literature procedures (J. Med. Chem., 2003, 46, 4050-4062). To a solution of compound 7-1 (1.0 g, 2.4 mmol) in acetonitrile (25 mL) was added l-(2-(pyrrolidin-3-yl)ethyl)piperidine trifluoroacetic acid (666 mg, 2.4 mmol) and potassium carbonate (660 mg, 2.4 mmol). The mixture was stirred at reflux for 16 h. After cooling to rt, the mixture was filtered and the filtrate was concentrated. The filtrate was then diluted with CH 2 Cl 2 (3OmL) and washed with saturated brine solution.
  • Compound 8-1 was prepared from similar procedures in Example 7. To a solution of 8-1 (90 mg, 0.17 mmol) in methanol (2 mL) was added 10% Pd/C (catalytic amount). The reaction was stirred under 1 arm of hydrogen for 16 h. The mixture was filtered and evaporated to afford 8-2 (70 mg, 0.16 mmol, 94%). Compound 8-2 was coupled with isopropylamine by using the same procedures in Example 7 to give 8-3. Data for 8-3: MS (ESI), m/z (assignment): 490/492 ([M+H] + , 100/35).
  • Table 1 shows compounds that exhibited IC5 0 less than 10 ⁇ M:

Abstract

Substituted pyridines, pyrimidines, pyrazines, pyridinones, pyrimidinones, pyrazinones and phenylacetamides useful in treating depression, stress and other disorders are disclosed. The compounds are of the formulae: (I).

Description

AZEVONE AND DIAZINONE V3 INHIBITORS FOR DEPRESSION AND STRESS DISORDERS
Field of the Invention
[0001] The invention relates to a chemical class of substituted pyridines, pyrimidines, pyrazines, pyridinones, pyrimidinones, pyrazinones and phenylacetamides useful in treating depression, stress and other disorders.
Background of the Invention
[0002] The hypothalamo-pituitary-adrenal (HPA) axis is the major stress axis in humans and other mammals. A variety of stressors (and multiple other classes of stimuli) cause release of the hormone ACTH (adrenocorticotropic hormone) from the anterior pituitary gland. ACTH enters the systemic circulation and acts on the adrenal cortex to promote synthesis and release of glucocorticoid hormone (the major endogenous glucocorticoid being Cortisol in humans and corticosterone in rodents). The glucocorticoids exert a broad spectrum of effects, the main purpose of which is to mobilize energy sources for successful responsiveness and eventual adaptation to the stressor.
[0003] Abnormally elevated HPA axis activity in man is associated with the development of a variety of psychiatric disturbances, some of which are stress- related in aetiology. Elevated Cortisol levels, which are indicative of HPA axis hyperactivity and loss of normal negative feedback regulatory processes, are a common finding in affective disorders and various other psychiatric disturbances, and are widely utilized as a diagnostic tool (Holsboer et al., Biol. Psych. 1986, 21, 601-611). It is generally considered that dysregulation of the HPA axis is a reflection of enhanced vulnerability and poor adaptation to chronic stress and that chronic stress therefore plays a major role in the development of affective illness (Sperry and Carlson, DSM-IV diagnosis to treatment, 2nd Edition, Taylor & Francis, 1996). This central concept is supported by experimental evidence utilizing animal models of chronic stress, where aberrant HPA function closely resembles that seen in clinical settings (De Goeij et al, Neuroendocrinology, 1991, 53, 150-159; Plotsky and Meaney, MoI. Brain Res. 1993, 18, 195-200).
[0004] The major secretogogues for ACTH in humans and rats are CRH (corticotropin releasing hormone) and AVP (arginine vasopressin). Within the HPA axis these peptide hormones are synthesized by the parvocellular neurones of the paraventricular nucleus (PVN) of the hypothalamus. The axons of these neurones project to the external zone of the median eminence, from where the hormone products enter the hypophysial portal system to bathe the corticotrope cells that manufacture ACTH. CRH and AVP act synergistically at the corticotrope to regulate ACTH secretion in both and in man.
[0005] The HPA axis is most potently activated by psychological stressors {i.e., those which require a cognitive assessment of the stimulus). The patterns of AVP and CRH release vary as a function of the type of stressor involved. Acute stress, whether physical or psychological, elicits rapid and robust CRH release. For several psychological stressors, however, chronic application elicits enhanced AVP storage in the median eminence, increased niRNA synthesis, and reduction in AVP neurosecretory granules, whereas similar markers of CRH synthesis and release are relatively unaffected. These findings, when considered together with clinical and experimental data indicating that stress enhances the number of PVN neurones co-expressing CRH and AVP, and that brain levels of AVP are elevated in patients suffering from affective disorders, show that AVP plays an important role as an ACTH secretagogue. Further, they show that chronic psychological stress is associated with a shift in emphasis from CRH to AVP-controlled HPA axis activity. Thus AVP plays a pivotal role in the genesis of the HPA hyperactivity documented in affective disorders. [0006] The actions of AVP at the pituitary cortocotrope are mediated by the vasopressin V3 (or VIb) receptor, which is known and has been cloned (human receptor: Sugimoto et al, J. Biol. Chem., 1994, 269, 27088-27092). A report of clinical studies in depressed patients in which blunted ACTH responses to CRH could be restored by concomitant administration of desmopressin (dDAVP, an AVP agonist with V3 affinity) confirms the involvement of the V3 receptor in depression (Scott and Dinan, Life Sciences, 1998, 62, 1985-1988). A study in rodents with non-selective peptide V3 antagonists indicates that the V3 receptor does play a functional role in control of pituitary ACTH release (Bernardini et al., Neuroendocrinology, 1994, 60, 503-508). Vasopressin antagonists are thus utilized to modulate and normalize pituitary ACTH release and subsequent HPA axis dysfunction in CNS disorders which are characterized by abnormal HPA axis negative feedback mechanisms.
[0007] Studies have indicated that V3 antagonists may be useful in the treatment of aggressive behavior [see Wersinger et al. MoI. Psychiatry 7. 975- 984 (2002); Blanchard et al. Pharmcol. Biochem. Behav. 80. 189-194 (2005); and Wersinger et al. Horm. Behav. 46, 638-645 (2004)]; insomnia in elderly patients [see Kalamatianos et al. J. Neuroendocrinol. 16, 493-501 (2004)]; cancer [see Dahia et al. J. Clin. Endocrin. Metab. 81. 1768-1771 (1996)]; Cushing's Disease [see Perraudin et al. J. Clin. Endocrin. Metab. 80. 2661-2667 (1995)]; pancreatic disease [see Folny et al. Am. J. Physiol. 285. E566-576 (2003)]; and to effect diuresis [see Chen et al. J. Neurosci. Res. 60. 761-766 (2000)].
[0008] In addition to the V3 receptor, vasopressin also activates peripheral receptors, i.e., the Via receptor, predominantly found on liver and vascular tissue and the V2 receptor, predominantly found on kidney tissue. Interaction at these receptors mediates the pressor and antidiuretic actions of AVP. [0009] Whilst there are several non-peptide low-molecular weight antagonists known which are selective for the Via or the V2 receptor (for a recent review see Freidinger and Pettibone, Medicinal Research Reviews, 1997, 17, 1-16), there are only a small number of non-peptide ligands known with selectivity for the V3 receptor (see for example, WO 01/55130 and WO 04/009585). There exists therefore a need for further non-peptide V3 selective antagonists which are both safe and effective.
Summary of the Invention
[0010] There are provided, in accordance with an embodiment of the invention, compounds of formulae:
Figure imgf000005_0001
Figure imgf000006_0001
In genera I and II, X4-X5 is chosen from CR4-CR5, N-CR5 and CR4-N; in genus in, one of X6, X7 and X8 is N and the other two are CR4 and CR5.
R1 is chosen from (C1-io)alkyl, (Cs.^cycloalkyl, [(C3-1o)cycloalkyl(Ci-2)alkyl], said (C1-1o)alkyl, (C3-1o)cycloalkyl, and [(C3-10)cycloalkyl (C1-2)alkyl] being optionally substituted with one or more halogens, (Q^alkoxy, (C2- 6)alkenyl, (C2-6)alkynyl, phenyl or benzyl; Ar is chosen from
(i) (C6-io)aryl, optionally substituted within 1-3 substituents selected from halogen, hydroxy, cyano, COOR5, NR6R7, phenyl, (C5-6)heteroaryl, (C1-
6)alkyl, (C3-6)cycloalkyl, (Ci-6)alkyloxy and (C3-6)cycloalkyloxy, said (Ci-
6)alkyl, (C3-6)cycloalkyl, (Ci-6)alkyloxy and (C3-6)cycloalkyloxy being optionally substituted with one or more halogens;
(ii) (C5-io)heteroaryl optionally substituted with a substituent selected from methyl,
Figure imgf000006_0002
or halogen; and
(iii) (C4-7)cycloalkyl; R4 and R5 are independently chosen from H, (Ci-6)alkyl, (Ci^alkyloxy or halogen, said (Ci^alkyl and (Ci^alkyloxy being optionally substituted with one or more halogens;
G is a linking moiety spanning 4 to 7 atoms between termini; and R8 and R9 are residues that, in combination, maintain the basicity of N. When X^-X5 is CR4-N and G is alkylene, R1 must be chosen from (Ci-6)alkyl, (C3-6)cycloalkyl and [(C3-6)cycloalkyl(Ci-2)alkyl]. When R1 is not restricted to this subset of values, G is a linking moiety incorporating at least one of (a) an sp2 hybridized carbon; or (b) a cyclic structure. Alternatively, when R8 and R9 together form a 4- to 7-membered nitrogenous heterocycle, G may additionally be -N(R10)-(C4-6)alkylene for all values of R1. R10 is H or (C1-6)alkyl.
[0011] A subgenus of compounds in accordance with embodiments of the invention is the genus I:
Figure imgf000007_0001
[0012] The genus I can be divided into three subgenera:
Figure imgf000007_0002
and
pyrazmones:
Figure imgf000008_0001
[0013] In another embodiment of the invention there are provided pharmaceutical formulations comprising a pharmaceutically acceptable carrier and a compound as described above.
[0014] In another embodiment of the invention there are provided methods for treating depression, stress disorders, aggressive behavior, insomnia in elderly patients, cancer, Cushing's Disease, and pancreatic disease and to effect diuresis using a compound as described above.
Detailed description of the Invention
[0015] In some embodiments of the invention, there are provided pyridines, pyrimidines, pyrazines, pyridinones, pyrimidinones, pyrazinones and phenylacetamides falling within a general formula
Figure imgf000008_0002
5 in which B represents a six-membered, planar carbocycle or planar nitrogen heterocycle.
[0016] As indicated above, G is a linking moiety spanning 4 to 7 atoms between termini. In other words, the -NR8R9 must be 4 to 7 atoms removed from the ring B. The precise constituents of G are not critical. Typically G will incorporate either an sp2 hybridized carbon or a cyclic structure. When R8 and R9 together form a 4- to 7-menibered nitrogenous heterocycle, G may be -N(R10)-(C4.6)alkylene. Thus, for example, G may be a (C4-C7)-alkylene in which one or more -CH2- may be replaced by -S-, -S(O)-, -SO2-, -O-, -C(=O)-, -CHOH-, -NH-, CHF, CF2, -CH(O-loweralkyl)-, -CH(O-loweracyl)-, -CH(loweralkyl)- or -C(loweralkyl)2-, with the provisos that (1) adjacent -CH2- residues cannot be replaced by -S-, -S(O)-, -SO2- or -0-; and (2) -S-, -S(O)-, - SO2-, -O- and -NH- residues cannot be separated only by a single carbon. G may also be an optionally substituted carbocycle or heterocycle, attached to the B ring and to -NR8R9 by a direct bond or by a C1-C5 alkylene chain. G may also be an optionally substituted nitrogenous heterocycle, attached to the B ring by a direct bond or by a C1-C5 alkylene chain; in this case a nitrogen of the nitrogenous heterocycle may correspond to -NR8R9 so that R9 becomes formally part of G. R and R may also be taken together and attach to G so as to form a nitrogen-containing heterocycle, e.g. a pyridine ring attached at one of the ring carbon atoms to the linker which links the pyridine ring to ring B. In such cases, there will be 4 to 7 atoms between the ring B and the carbon atom of the nitrogen-containing heterocycle.
[0017] The residues R8 and R9 must maintain the basicity of N. For example, alkyl residues of various sorts are within the invention; alkylene and similar residues (e.g. alkylene with heteroatom interruption) that tie the nitrogen into a ring are within the invention. Even residues that introduce aromaticity are tolerated, as long as the nitrogen remains basic (e.g. pyridine). Acyl residues (e.g. R8 = acetyl), which destroy the basicity of the nitrogen, are outside the invention. Under certain circumstances, one or both of R8 and R9 may be hydrogen. These concepts are explained more fully in the text and examples below.
[0018] A genus in accordance with some embodiments of the invention comprises pyridinones, pyrimidinones and pyrazinones that fall within the general formula I above:
Figure imgf000010_0001
. In these compounds, X4-
X5 is CR4-CR5, N-CR5 or CR4-N.
[0019] Examples of subgenera in accordance with embodiments of the invention include the subgenus in which -NR8R9 is a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings, preferably a piperidine or morpholine, and G is a (Cs.^hydrocarbon chain attached to the pyridinone, pyrimidinone or pyrazinone through an amide or amine:
Figure imgf000010_0002
wherein E is (C2-1o)hydrocarbon; and
Figure imgf000011_0001
— is a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings. The nitrogenous heterocycle may be substituted, for example, 4-hydroxypiperidin-l-yl, 4-hydroxy-4- methylpiperidin-1-yl and 4,4-dimethylpiperidin-l-yl. The (C2-io)hydrocarbon may be straight chain, branched or cyclic as long as the 4-7 atom spacing between -NR8R9 and the pyridinone, pyrimidinone or pyrazinone ring is maintained. Examples of species in this subgenus include:
Figure imgf000012_0001
and
Figure imgf000012_0002
[0020] A further example of a subgenus in accordance with embodiments of the invention is the subgenus in which -NR8R9 is attached via a direct bond or (Ci- 6)hydrocarbon, J, to a single ring carbocycle or heterocycle of 4 to 7 atoms or a
two ring carbocycle or heterocycle of 9 to 13 atoms,
Figure imgf000012_0003
. The carbocycle or heterocycle
Figure imgf000013_0001
is directly attached to the pyridinone, pyrimidinone or pyrazinone:
Figure imgf000013_0002
nitrogenous heterocycle (e.g. oxadiazolyl, pyrrolidinyl and piperidinyl) and J is methylene, ethylene or propylene, and in some embodiments -NR8R9 is chosen from piperidine, morpholine and -N[C1-3alkyl]2. Examples of species in this subgenus include:
Figure imgf000013_0003
Figure imgf000014_0001
[0022] A further example of a subgenus in accordance with embodiments of the invention is the subgenus in which R is alkylene or similar residue (e.g. alkylene with heteroatom interruption) "tied back" into a nitrogen that is directly attached to the pyridinone, pyrimidinone or pyrazinone ring:
Figure imgf000014_0002
wherein
Figure imgf000014_0003
is a nitrogenous single ring heterocycle of 6 to 8 atoms or a two ring heterocycle of 9 to 13 atoms in which the nitrogen labeled b is the nitrogen of claim 1 and the nitrogen labeled a is subsumed in the definition of G. In one
embodiment,
Figure imgf000014_0004
is a hexahydro-l,4-diazepine ring. An example is
Figure imgf000015_0001
A further example of a subgenus in accordance with embodiments of the invention is the subgenus in which R9 is alkylene or similar residue (e.g. alkylene with heteroatom interruption) "tied back" into a chain that is attached to the pyridinone, pyrimidinone or pyrazinone ring through an amine or amide linkage. These compounds may be thought of as a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings attached to the pyridinone, pyrimidinone or pyrazinone ring through an alkylene chain, an amine or an amide linkage:
Figure imgf000015_0002
Figure imgf000016_0001
in one or two rings; and R8 is C1-10 hydrocarbon. In certain embodiments,
Figure imgf000016_0002
is a piperidine ring and R is methyl. An example is:
Figure imgf000016_0003
This particular subgenus also includes compounds of the formulae
Figure imgf000017_0001
in which R1 is C3-C6 alkyl. An example is
Figure imgf000017_0002
[0023] As explained above, R8 and R may be taken together and attach to G so as to form a nitrogen-containing heterocycle. There are three genera that, while conceptually subgenera of the genus I, may not be sensu strictu within the Markush parent structure set forth above for I. These are the pyridinones, pyrimidinones and pyrazolones formulae:
Figure imgf000018_0001
in which Pyr represents imidazole, pyridine attached through a carbon, substituted imidazole or substituted pyridine attached through a carbon. An example is:
Figure imgf000018_0002
[0024] Among the foregoing genera, R1 may be chosen from C3-6alkyl and cycloalkyl and Ci-3 alkyl substituted with phenyl, methoxy or alkynyl. For example R1 may be t-butyl, isopropyl, cyclopentyl, α-methylbenzyl, methoxypropyl or propargyl. Ar may be chosen from phenyl and phenyl substituted with halogen (e.g. chloro and fluoro), Ci-2alkyl, (e.g. methyl) trifluoromethyl, C1-3 alkyloxy (e.g. methoxy), C1-4 cycloalkyloxy or trifluoromethoxy. In some embodiments, Ar is a 3-substituted phenyl ring, for example a substituted phenyl ring selected from 3-chlorophenyl, 3- fiuorophenyl, 3-methoxyphenyl, 3-trifluoromethoxyphenyl, 3-chloro-4- fluorophenyl, 4-fluoro-3-methoxyphenyl and 3,5-dimethoxyphenyl.
[0025] Since the compounds in accordance with embodiments of the invention all contain a basic nitrogen, they may be presented as salts. In the claims, reference to the compound includes its salts. The term "pharmaceutically acceptable salt" refers to salts whose counter ion derives from pharmaceutically acceptable non-toxic acids and bases. Suitable pharmaceutically acceptable base addition salts for the compounds of the present invention include inorganic acids, and organic acids. Examples include acetate, benzenesulfonate (besylate), benzoate, bicarbonate, bisulfate, carbonate, camphorsulfonate, citrate, ethanesulfonate, fumarate, gluconate, glutamate, glycolate, bromide, chloride, isethionate, lactate, maleate, malate, mandelate, methanesulfonate, mucate, nitrate, pamoate, pantothenate, phosphate, succinate, sulfate, tartrate, trifluoroacetate, p-toluenesulfonate, acetamidobenzoate, adipate, alginate, aminosalicylate, anhydromethylenecitrate, ascorbate, aspartate, calcium edetate, camphorate, camsylate, caprate, caproate, caprylate, cinnamate, cyclamate, dichloroacetate, edetate (EDTA), edisylate, embonate, estolate, esylate, fluoride, formate, gentisate, gluceptate, glucuronate, glycerophosphate, glycolate, glycollylarsanilate, hexylresorcinate, hippurate, hydroxynaphthoate, iodide, lactobionate, malonate, mesylate, napadisylate, napsylate, nicotinate, oleate, orotate, oxalate, oxoglutarate, palmitate, pectinate, pectinate polymer, phenylethylbarbiturate, picrate, pidolate, propionate, rhodanide, salicylate, sebacate, stearate, tannate, theoclate, tosylate, and the like. When the compounds contain an acidic residue, suitable pharmaceutically acceptable base addition salts for the compounds of the present invention include ammonium, metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, N5N'- dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. Other base addition salts includes those made from: arecoline, arginine, barium, benethamine, benzathine, betaine, bismuth, clemizole, copper, deanol, diethylamine, diethylaminoethanol, epolamine, ethylenediamine, ferric, ferrous, glucamine, glucosamine, histidine, hydrabamine, imidazole, isopropylamine, manganic, manganous, methylglucamine, morpholine, morpholineethanol, n- ethylmorpholine, n-ethylpiperidine, piperazine, piperidine, polyamine resins, purines, theobromine, triethylamine, trimethylamine, tripropylamine, trolamine, and tromethamine.
Definitions
[0026] Throughout this specification the terms and substituents retain their definitions.
Alkyl is intended to include linear, branched, or cyclic hydrocarbon structures and combinations thereof. When not otherwise restricted, the term refers to alkyl of 20 or fewer carbons. Lower alkyl refers to alkyl groups of 1-6 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s-and t-butyl and the like. Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of 3-8 carbon atoms. Examples of cycloalkyl groups include c-propyl, c-butyl, c-pentyl, norbornyl, adamantyl and the like. In accordance with standard nomenclature, the term "alkylene" applies to alkyl residues having two points of attachment. For example, propylene refers to -CH2CH2CH2-.
[0027] The term "hydrocarbon" includes alkyl, cycloalkyl, alkenyl, alkynyl, aryl and combinations thereof. Examples include benzyl, phenethyl, cyclohexylmethyl, camphoryl and naphthylethyl.
[0028] Alkoxy or alkoxyl refers to groups of 1-8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to four carbons.
[0029] Oxaalkyl refers to alkyl residues in which one or more carbons (and their associated hydrogens) have been replaced by oxygen. Examples include methoxypropoxy, 3,6,9-trioxadecyl and the like. The term oxaalkyl is intended as it is understood in the art [see Naming and Indexing of Chemical Substances for Chemical Abstracts, published by the American Chemical Society, ^fI 96, but without the restriction of Tfl27(a)], i.e. it refers to compounds in which the oxygen is bonded via a single bond to its adjacent atoms (forming ether bonds). Similarly, thiaalkyl and azaalkyl refer to alkyl residues in which one or more carbons have been replaced by sulfur or nitrogen, respectively. Examples include ethylaminoethyl and methylthiopropyl.
[0030] Acyl refers to groups of 1-8 carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality. One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl. Examples include formyl, acetyl, propionyl, isobutyryl, ^-butoxycarbonyl, benzoyl, benzyloxycarbonyl and the like. Lower-acyl refers to groups containing one to four carbons.
[0031] Aryl and heteroaryl refer to aromatic or heteroaromatic rings, respectively, as substituents. Heteroaryl contains one, two or three heteroatoms selected from O, N, or S. Both refer to monocyclic 5- or 6-membered aromatic or heteroaromatic rings, bicyclic 9- or 10-membered aromatic or heteroaromatic rings and tricyclic 13- or 14-membered aromatic or heteroaromatic rings. Aromatic 6-14-membered carbocyclic rings include, e.g., benzene, naphthalene, indane, tetralin, and fluorene and the 5-10-membered aromatic heterocyclic rings include, e.g., imidazole, pyridine, indole, thiophene, benzopyranone, thiazole, furan, benzimidazole, quinoline, isoquinoline, quinoxaline, pyrimidine, pyrazine, tetrazole and pyrazole.
[0032] Arylalkyl means an alkyl residue attached to an aryl ring. Examples are benzyl, phenethyl and the like.
[0033] Substituted alkyl, aryl, cycloalkyl, heterocyclyl etc. refer to alkyl, aryl, cycloalkyl, or heterocyclyl wherein up to three H atoms in each residue are replaced with halogen, haloalkyl, hydroxy, loweralkoxy, carboxy, carboalkoxy (also referred to as alkoxycarbonyl), carboxamido (also referred to as alkylaminocarbonyl), cyano, carbonyl, nitro, amino, alkylamino, dialkylamino, mercapto, alkylthio, sulfoxide, sulfone, acylamino, amidino, phenyl, benzyl, heteroaryl, phenoxy, benzyloxy, or heteroaryloxy.
[0034] The term "halogen" means fluorine, chlorine, bromine or iodine.
[0035] In the characterization of the variables, it is recited that various R-groups may form rings or heterocycles. For example, R8 and R9 together form a 4- to 7-membered nitrogenous heterocycle. It is intended that these rings may exhibit various degrees of unsaturation, may include heteroatoms and may be substituted with lower alkyl or alkoxy.
[0036] It will be recognized that the compounds of this invention can exist in radiolabeled form, i.e., the compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Radioisotopes of hydrogen, carbon, phosphorous, fluorine, and chlorine include 3H, 14C, 35S, 18F, and 36Cl, respectively. Compounds that contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of embodiments of the invention. Tritiated, i.e. 3H, and carbon-14, i.e., 14C, radioisotopes are particularly known for their ease in preparation and detectability. Radiolabeled compounds of this invention can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabeled compounds can be prepared by carrying out the procedures disclosed in the Examples and Schemes by substituting a readily available radiolabeled reagent for a non-radiolabeled reagent. Radiolabeled compounds are useful in screens for V3 agonists and antagonists.
[0037] The compounds described herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms. Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. Included in embodiments of the present invention are all such possible isomers, as well as their racemic and optically pure forms. Optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
[0038] The graphic representations of racemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are taken from Maehr L Chem. Ed. 62, 114-120 (1985): solid and broken wedges are used to denote the absolute configuration of a chiral element; wavy lines and single thin lines indicate disavowal of any stereochemical implication which the bond it represents could generate; solid and broken bold lines are geometric descriptors indicating the relative configuration shown but denoting racemic character; and wedge outlines and dotted or broken lines denote enantiomerically pure compounds of indeterminate absolute configuration.
[0039] The abbreviations Me, Et, Ph, Tf, Ts and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, toluenesulfonyl and methanesulfonyl respectively. The following abbreviations and terms have the indicated meanings throughout:
abs = absolute
Ac acetyl
ACN acetonitrile
Bu butyl c- = cyclo
CDI carbonyldiimidazole cone. = concentrated
DCM dichloromethane = methylene chloride = CH2Cl2
DIC diisopropylcarbodiimide
DMAP 4-N,N-dimethylaminopyridine
DMF N,N-dimethylformamide
DMSO dimethyl sulfoxide
DPPA diphenylphosphoryl azide dppf bisdiphenylphosinoferrocene
EDC, EDCI = 1 -ethyl-3 -(3 '-dimethylaminopropyl)carbodiimide
Et ethyl
FCC flash column chromatography
GC gas chromatography HOAc = acetic acid
HOBt = hydroxybenzotriazole i- = iso-
IBCF = isobutylchloroformate
IPA = isopropyl alcohol
Me = methyl
MP = macroporous
NMM = N-methylmorpholine
NMO = N-methylmorpholine oxide
Ph = phenyl
PhOH = phenol ppt. = precipiate
PPTS = pyridinium p-toluenesulfonate
Pr = propyl
PS = polystyrene rt = room temperature sat'd = saturated s- = secondary t- = tertiary
TBDMS = t-butyldimethylsilyl
TEA = triethylamine
TFA = trifluoroacetic acid
THF = tetrahydrofuran
TLC = thin-layer chromatography
TMS = trimethylsilyl tosyl = p-toluenesulfonyl
A comprehensive list of abbreviations utilized by organic chemists (i.e. persons of ordinary skill in the art) appears in the first issue of each volume of the
Journal of Organic Chemistry. The list, which is typically presented in a table entitled "Standard List of Abbreviations" is incorporated herein by reference. [0040] While it may be possible in accordance with some embodiments of the invention for the compounds to be administered as the raw chemical, in other embodiments the compounds are presented in a pharmaceutical composition. In accordance with an embodiment of the invention, there is provided a pharmaceutical composition comprising a compound as described herein or a pharmaceutically acceptable salt or solvate thereof, together with one or more pharmaceutically carriers thereof and optionally one or more other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
[0041] The formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular), rectal and topical (including dermal, buccal, sublingual and intraocular) administration. The most suitable route may depend upon the condition and disorder of the recipient. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods of treatment in accordance with embodiments of the invention include the step of bringing into association a compound in accordance with embodiments of the invention or a pharmaceutically acceptable salt or solvate thereof ("active ingredient") with the carrier, which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
[0042] Formulations in accordance with embodiments of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in- water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
[0043] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein.
[0044] The pharmaceutical compositions may include a "pharmaceutically acceptable inert carrier", and this expression is intended to include one or more inert excipients, which include starches, polyols, granulating agents, macrocrystalline cellulose, diluents, lubricants, binders, disintegrating agents, and the like. If desired, tablet dosages of the disclosed compositions may be coated by standard aqueous or nonaqueous techniques, "Pharmaceutically acceptable carrier" also encompasses controlled release means.
[0045] Compositions in accordance with embodiments of the present invention may also optionally include other therapeutic ingredients, anti-caking agents, preservatives, sweetening agents, colorants, flavors, desiccants, plasticizers, dyes, and the like. Any such optional ingredient must, of course, be compatible with the compound of the invention to insure the stability of the formulation.
[0046] The dose range for adult humans is generally from 0.005 mg to 10 g/day orally. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of a compound or mixture of compounds in accordance with embodiments of the invention which is or are effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg. The precise amount of compound or compounds administered to a patient will be the responsibility of the attendant physician. However, the dose employed will depend on a number of factors, including the age and sex of the patient, the precise disorder being treated, and its severity.
[0047] Compounds in accordance with embodiments of the present invention can be prepared by the following methods:
Example 1. Synthesis of Intermediate 1-4
Figure imgf000028_0001
Figure imgf000028_0002
Minor
Figure imgf000028_0003
Step l:
[0048] 3-methoxyacetophenone (18.0 g, 120.0 mmol) and dimethylformamide dimethyl acetal (37.5 g, 315 mmol) were combined neat in a microwave reaction vessel and irradiated with microwave energy to 2000C for 10 min. The reaction mixture was concentrated in vacuo and purified by flash column chromatography (SiO2; elution with 7:3 hexane/EtOAc then 1:4.5:4.5
MeOH/EtOAc/hexane) collecting to provide 26.Og (100%) of the enaminone 1-
1 as a dark, red-orange oil.
Data for 1-1: 1H NMR (300 MHz, CDC13): δ 7.75 (d, IH), 7.41 (m, 2H), 7.23
(d, IH), 6.95 (ddd, IH), 5.64 (d, IH), 3.80 (s, 3H), 3.08 (br s, 3H), 2.87 (br s,
3H); MS (ESI), m/z (relative intensity, assignment) 206.1 (100, [M+H]+).
[0049] To a solution of 1-1 (26.8 g, 130.5 mmol) in dimethylformamide (250 mL) were added sodium hydride (60%, 6.27 g, 261 mmol) and cyanoacetamide (11.0 g, 130.5 mmol). The mixture was then stirred at 1050C for 2h. This was then concentrated in vacuo and the crude residue taken up in water (600 mL). The pH of the solution was adjusted with acetic acid, then warmed to 7O0C for 15 min and the resultant yellow ppt. collected by filtration. This was washed with hot water (3 X 150 mL) followed by cold methanol (1 X 500 mL) and dried overnight in a vacuum oven to provide 28.4 g (91%) 1-2 as a tan solid. Data for 1-2: 1H NMR (300 MHz, DMSOd6): δ 12.72 (br s, IH), 8.20 (d, IH), 7.50 -7.30 (m, 3H), 7.12 (m, IH), 6.80 (br d, IH), 3.82 (s, 3H); MS (ESI), m/z (relative intensity, assignment) 227.2 (100, [M+H]+).
Step 3:
[0050] 1-2 (28.4 g, 118.2 mmol) and potassium carbonate (49 g, 355mmol) were combined in anhydrous acetone (40OmL) and stirred. To the stirred solution was added allyl bromide (20.5 mL; 236 mmol) and the mixture was heated to reflux with stirring for 16 h. This was cone, in vacuo and partitioned between water and DCM. The organic phase was dried over MgSO4, filtered and cone, in vacuo to provide 32 g (100%) of crude product as a mixture of 1- 3a (major) and l-3b (minor). The crude residue was used in the next reaction without further purification.
Data for l-3a: 1H NMR (300 MHz, CDC13): δ 7.92 (d, IH), 7.63-7.57 (m, 2H), 7.45-7.37 (m, 2H), 7.03 (m, IH), 6.14 (ddt, IH), 5.50 (d, IH), 5.32 (d, IH), 5.07 (d, 2H), 3.89 (s, 3H); MS (ESI), m/z (relative intensity, assignment) 267.1 (100, [M+H]+).
Step 4:
[0051] To a solution of l-3a and l-3b from the previous step (10.5 g, 36 mmol) in DCM (150 mL) was added bis(acetonitrile) palladium(II) chloride (2.0 g, 3.6 mmol) and the reaction mixture stirred for 4 h. This was then filtered through a pad of Celite and concentrated in vacuo. The crude residue was purified by
FCC (SiO2; elution with 2:1 hexanes/EtOAc) to afford 5.0 g (52%) of 1-4 as a yellow solid.
Data for 1-4: 1H NMR (300 MHz, CDC13): δ 7.82 (d, IH), 7.39 (dd, IH), 7.04
(ddd, IH), 6.90 (ddd, IH), 6.85 (dd, IH), 6.19 (d, IH), 5.85 (ddt, IH) 5.19 (d,
IH), 4.92 (d, IH), 4.53 (d, 2H), 3.83 (s, 3H); MS (ESI), m/z (relative intensity, assignment) 267.1 (100, [M+H]+).
Example 2. Preparation of 2-4
Figure imgf000030_0001
Figure imgf000031_0001
94% for 2 steps
HOBt, PS-carbodiimide resin, DCM, /-PrNH2; MP-carbonate resin
Figure imgf000031_0002
Step 1:
[0052] To a solution of 1-4 (1 g; 3.75 mmol) in 80% EtOH (aq) (10 mL) was added KOH (843 mg; 15.02 mmol). The reaction mixture was then heated to reflux for 16 h. The mixture was cooled to room temperature and partitioned between H2O (100 mL) and EtOAc (50 mL). The aq. phase was acidified to pH 3 with 2 N HCl (aq) and extracted with EtOAc (3 X 50 mL). The combined organic phases were dried (MgSO4), filtered and cone, in vacuo giving 990 mg (93%) of 2-1.
Data for 2-1: 1H NMR (300 MHz, CDC13): δ 14.3 (br s, IH), 8.55 (d, IH), 7.41 (dd, IH), 7.08 (dd, IH), 6.92 (d, IH), 6.87 (d, IH), 6.50 (d, IH), 5.89 (ddt, IH), 5.23 (d, IH), 4.92 (d, IH), 4.62 (d, 2H), 3.82 (s, 3H); MS (ESI), m/z (relative intensity, assignment) 286.1 (79, [M+H]+).
Step 2:
[0053] To a solution of 2-1 (223 mg; 0.78 mmol) in THF (5 mL) was added CDI (253 mg; 1.56 mmol). The reaction mixture was heated to 5O0C with stirring for 2 h. The mixture was then cooled to 230C and 3-(piperidin-l- yl)propan-l -amine (0.7 mL; 3.9 mmol) was added. This was stirred at 230C for 3 h. The mixture was then partitioned between sat'd NaHCO3 (aq) (50 mL) and
EtOAc (3 X 30 mL). The combined organic phases were dried (Na2SO4), filtered and cone, in vacuo. The crude residue was purified by FCC (SiO2; elution with 10% MeOH/DCM w/ 0.5% NH4OH (aq)) giving 97 mg (30%) of semi-pure 2-2.
Data for 2-2: MS (ESI), m/z (relative intensity, assignment) 410.2 (100,
[M+H]+).
Step 3:
[0054] The semi-pure amine 2-2 from step 2 (97 mg; 0.24 mmol) was taken up in DCM and treated with excess TFA then cone, in vacuo to protect the amine as a TFA salt. This was then dissolved in DCM (3 mL) and MeOH (1 mL) and cooled to -780C. O3 was bubbled through until a blueish color persisted for 5 min. A stream of Ar was then passed through the solution to remove excess O3. Methyl sulfide (0.1 mL; 1.2 mmol) was added and the resultant mixture warmed to 230C and stirred for 16 h. The mixture was cone, in vacuo and the resultant aldehyde used crude in the next reaction.
Step 4:
[0055] The crude aldehyde from the previous step was taken up in £-BuOH (4 mL) and H2O (1 mL). To this was added NaH2PO4 (101 mg; 0.84 mmol), 2- methyl-2-butene (2.0 M in THF; 0.72 mL; 1.44 mmol) and NaClO2 (80%; 35 mg; 0.312 mmol). The reaction mixture was stirred at 23°C for 1.5 h. The mixture was then cone, in vacuo and the crude residue taken up in H2O (3 mL). This was acidified to pH 2 with 2 N HCl (aq). The mixture was then applied to a column packed with Dowex 50WX4-400 H+ ion exchange resin (~ 10 g) and eluted with 4: 1 H2O/ACN until the eluent became neutral to pH paper. This was then eluted with 4:1 H2O/ACN containing 10% cone. NH4OH (aq). The desired fractions containing product were" combined and cone, in vacuo giving 97 mg (94% for 2 steps) of carboxylic acid 2-3.
Data for 2-3: 1H NMR (300 MHz, CD3OD): δ 8.42 (d, IH), 7.40 (dd, IH), 7.12 - 7.03 (m, 3H), 6.45 (d, IH), 4.45 (br s, 2H), 3.82 (s, 3H), 3.53 (t, 2H), 3.25 - 3.00 (m, 6H), 2.05 (pentet, 2H), 1.83 (m, 4H), 1.65 (m, 2H); LC/MS, m/z (relative intensity, assignment) 428.1 (100, [MfH]+).
Step 5:
[0056] A mixture of the acid 2-3 (19 mg; 0.044 mmol), HOBt (7 mg; 0.0503 mmol) and PS-carbodiimide resin (Argonaut; 1.2 mmol/g; 49 mg; 0.0592 mmol) in DCM (1 mL) was stirred for 5 min. To this was added /-PrNH2 (30 μL; 0.0296 mmol) and stirring continued for 16 h. To this was then added MP- carbonate resin (Argonaut; 2.9 mmol/g; 210 mg; 0.609 mmol) and stirring continued for 2 h. This was then filtered and cone, in vacuo giving 15.1 mg (100%) of 2-4.
Data for 2-4: 1H NMR (300 MHz, CDCl3): δ 9.63 (br t, IH, amide NH), 8.57 (d, IH), 7.38 (dd, IH), 7.02 (m, 3H), 6.41 (d, IH), 5.65 (br d, IH, amide NH), 4.45 (s, 2H), 4.08 (m, IH), 3.82 (s, 3H), 3.48 (q, 2H), 2.38 (m, 6H), 1.81 (pentet, 2H), 1.58 (m, 4H), 1.42 (m, 2H), 1.16 (d, 6H); MS (ESI)5 m/z (relative intensity, assignment) 469.2 (100, [M+H]+).
Example 3. Preparation of 3-3
-780C; 2-methyl-2-butene,
Figure imgf000033_0001
resin, DCM, resin
Figure imgf000034_0001
Figure imgf000034_0002
Step 1:
[0057] To a solution of acid 2-1 (442 mg; 1.55 mmol) in THF (10 mL) was added CDI (0.51 g; 3.10 mmol). The reaction mixture was heated to 5O0C and stirred for 1.5 h. The mixture was cooled, cone, in vacuo and the crude residue partitioned between EtOAc (100 mL) and H2O (50 mL). The organic phase was dried (Na2SO4), filtered and cone, in vacuo.
NaH (60%; 65 mg; 1.63 mmol) was added to a mixture of N'-hydroxy-3- (piperidin-l-yl)propanamidine (319 mg; 1.86 mmol) and 4 A molecular sieves (1 scoop) in DMF (10 mL) and this was stirred for 30 min at 23°C. To this was added the imidazolide from above via cannula as a soln. in DMF (3 mL w/ 2 mL rinse). The reaction mixture was heated to 8O0C for 3 h. The mixture was cooled and partitioned between H2O (100 mL) and 3:1 DCM/f-PrOH (3 X 40 mL). The combined organic phases were washed with brine (1 X 50 mL), dried (K2CO3), filtered and cone, in vacuo. The crude residue was purified by FCC (SiO2; elution with 10% MeOH/DCM w/ 0.5% cone. NH4OH (aq)) giving 159 mg (24% from 2-1) of oxadiazole 3-1. Data for 3-1: LC/MS, m/z (relative intensity, assignment) 421.2 (100, [M+H]+).
Step 2 and 3:
[0058] Carboxylic acid 3-2 was prepared from alkene 3-1 (159 mg; 0.378 mmol) using the general procedures analogous to those described in Example 2, Steps 3 and 4. This afforded 130 mg (78% for 2 steps from 3-1) of acid 3-2. Data for 3-2: MS (ESI), m/z (relative intensity, assignment) 439.2 (100, [MB-H]+).
Step 4:
[0059] 3-3 was prepared from acid 3-2 (25 mg; 0.057 mmol) and Z-PrNH2
(0.038 mmol) using the same general procedure as described in Example 2, step
5. The product was further purified by prep. HPLC giving 6.8 mg (21%) 3-3 as a TFA salt.
Data for 3-3 (TFA salt): 1H NMR (300 MHz, CD3OD): δ 8.58 (d, IH), 7.44
(dd, IH), 7.12 (dd, IH), 7.04 (m, 2H), 6.61 (d, IH), 4.62 (s, 2H), 4.03 (t, 2H),
3.92 (m, IH, overlap with peak at 3.90), 3.90 (t, 2H, overlap with peak at 3.92),
3.81 (s, 3H), 3.61 (m, 4H), 2.13 (m, 2H), 1.93 (m, 2H), 1.76 (m, 2H); MS (ESI), m/z (relative intensity, assignment) 480.3(100, [M+H]+).
6 022025
Example 4. Preparation of 4-5.
(6:1) NaH2PO4, 2-methyl-2-butene,
Figure imgf000036_0001
Figure imgf000036_0002
reflux
Figure imgf000036_0003
Figure imgf000036_0004
Step 1:
[0060] To a solution of 1-4 (4.5 g; 16.9 mmol) in THF (60 mL) and H2O (10 mL) was added NMO (2.57 g; 21.97 mmol) and OsO4 (2.5 wt.% in if-BuOH; 2.12 mL; 0.169 mmol). The reaction mixture was stirred for 16 h, then 10% Na2S2O3 (aq) (5 mL) was added and stirred for 5 min. Three scoops of Celite were then added and the mixture filtered through a plug of Celite and cone, in vacuo. The crude residue was taken up in EtOAc (200 mL) and washed with 10% Na2S2O3 (aq) (1 X 50 mL), 1 N HCl (aq) (1 X 50 mL) and brine (1 X 50 mL). The organic phase was dried (Na2SO4), filtered and cone, in vacuo. The desired diol was used without further purification in the next step. Step 2:
[0061] The product diol from Step 1 was taken up in EtOH (60 niL) and to this was added NaIO4 (6.51 g; 30.43 mmol) dissolved in H2O (15 niL). The reaction mixture was stirred for 1 h 40 min. This was then filtered through a pad of
Celite with EtOH rinses and cone, in vacuo. The crude residue was partitioned between EtOAc (200 mL) and H2O (100 mL). The organic phase was dried
(Na2SO4), filtered and cone, in vacuo. The desired aldehyde 4-1 was used without further purification in the next step.
Data for 4-1: MS (ESI), m/z (relative intensity, assignment) 269.1 (100,
[M+H]+).
Step 3:
[0062] To a solution of the crude aldehyde from step 2 (~16.9 mmol) in t- BuOH (120 mL) and H2O (30 mL) was added 2-methyl-2-butene (2.0 M in THF; 55.0 mL; 110 mmol), NaH2PO4 (7.4 g; 61.6 mmol) and NaClO2 (80%; 2.21 g; 19.53 mmol). The reaction mixture was stirred at 23°C for 1.5 h. The mixture was then cone, in vacuo and the crude residue taken up in EtOAc (200 mL) and extracted with 1 N NaOH (aq) (1 X 100 mL then 2 X 50 mL). The combined aqueous phases were then acidified with cone. HCl (aq) giving a cloudy white precipitate. This was extracted with DCM (3 X 100 mL). The combined organic phases were washed with brine (1 X 100 mL), dried (Na2SO4), filtered and cone, in vacuo giving 3.42 g (71% for 3 steps from 1-4) of the acid 4-2.
Data for 4-2: 1H NMR (300 MHz, CDCl3 w/ drop of CD3OD): δ 7.89 (d, IH), 7.41 (dd, IH), 7.05 (dd, IH), 6.94 (dd, IH), 6.90 (dd, IH), 6.30 (d, IH), 4.55 (s, 2H), 3.82 (s, 3H); MS (ESI), m/z (relative intensity, assignment) 285.0 (100, [M+H]+). Step 4:
[0063] To a solution of 4-2 (1.7 g; 6.00 mmol) and ^-BuNH2 (0.75 mL; 7.18 mmol) in DCM (20 mL) was added EDCI (1.27 g; 6.6 mmol) and HOBt (892 mg; 6.6 mmol). The reaction mixture was stirred for 16 h. This was then diluted with EtOAc (200 mL) and washed with 1 N HCl (aq) (1 X 50 mL), sat. NaHCO3 (aq) (1 X 50 mL) and brine (1 X 50 mL). The organic phase was dried (Na2SO4), filtered and cone, in vacuo. The crude residue was purified by FCC (SiO2; elution with 1:1 EtOAc/hexanes) to afford 1.62 g (79%) of amide 4-3. Data for 4-3: 1H NMR (SOO MHZ5 CDCI3): δ 7.83 (d, IH), 7.38 (dd, IH), 7.10 - 6.96 (m, 3H), 6.25 (d, IH), 6.00 (br s, IH, amide NH), 4.42 (s, 2H), 3.82 (s, 3H), 1.33 (s, 9H); MS (ESI), m/z (relative intensity, assignment) 339.8 (28,
Figure imgf000038_0001
Step 5:
[0064] To a suspension of 4-3 (1.62 g; 4.77 mmol) in MeOH (15 mL) was added NH2OH*HC1 (531 mg; 7.64 mmol) and NaOMe (850 mg; 15.74 mmol).
The reaction mixture was heated to reflux for 6 h. This was then cooled to room temperature and quenched with sat. NH4Cl (aq) (50 mL). Extracted with
3:1 DCM/z-PrOH (3 X 40 mL). The combined organic phases were washed with brine (1 X 50 mL), dried (Na2SO4), filtered and cone, in vacuo giving 1.6 g
(90%) of amidoxime 4-4 which was used without further purification in the next step.
Data for 4-4: LC/MS, m/z (relative intensity, assignment) 373.0 (100, [MH-H]+).
Step 6:
[0065] To a solution of amidoxime 4-4 (1.5 g; 4.03 mmol) in DMF (40 mL) was added NaH (60%; 145 mg; 4.83 mmol). The resultant mixture was stirred at 230C for 30 min. To this was added ethyl 3-(piperidin-l-yl)propanoate (1.49 g; 8.06 mmol) via cannula as a solution in DMF (5 mL). The resultant mixture was heated to 8O0C for 1.5 h. The mixture was then cooled and partitioned between H2O (400 mL) and EtOAc (3 X 100 mL). The combined organic phases were dried (Na2SO4), filtered and cone, in vacuo. The crude residue was purified by FCC (SiO2; elution with 5% MeOH/DCM then 10% MeOH /DCM) giving 430 mg of semipure product. This was further purified by prep. HPLC giving the TFA salt of 4-5. The TFA salt of 4-5 was treated with 10% HCl/EtOH (excess) and cone, in vacuo. This was repeated twice more to convert the TFA salt to an HCl salt. The material was then triturated with Et2O giving an off-white solid which was dried in vacuo at 8O0C for 16 h. This afforded 223 mg (11%) of 4-5 as an HCl salt.
Data for 4-5 (HCl salt): 1H NMR (300 MHz, CD3OD): δ 8.39 (d, IH), 7.69 (br s, IH) 7.41 (dd, IH), 7.12 - 6.99 (m, 3H), 6.48 (d, IH), 4.60 (br s, 2H), 3.81 (s, 3H), 3.75 (t, 2H, partially obscured by peak at 3.70), 3.70 (m, 2H, partially obscured by peak at 3.75), 3.56 (t, 2H), 3.10 (br t, 2H), 2.05-1.75 (m, 5H), 1.59 (m, IH), 1.30 (s, 9H); LC/MS, m/z (relative intensity, assignment) 494.1 (100, [M+H]+).
Example 5. Preparation of 5-8.
Figure imgf000039_0001
Figure imgf000040_0001
Step 1:
[0066] To a solution of 3-chlorobenzonitrile (50 g, 363 mmol) in anhydrous
EtOH (500 mL), cooled to O0C in an ice bath, was bubbled HCl (g) through a gas dispersion tube for approximately 20 minutes until the solution was saturated. The resulting reaction mixture was stirred at room temperature for 16 h. Volatiles were removed in vacuo and the residue was triturated with anhydrous ether (~200mL). The white solid was collected by filtration and dried in vacuo overnight yielding 80 g (100%) of 5-1.
Data for 5-1: 1H NMR (300 MHz5 /-DMS0): δ 12.0-11.8 (br s, 1 H), 8.22-
8.17 (t, 1 H), 8.10-8.04 (dt, 1 H), 7.90-7.85 (dt, 1 H), 7.71-7.64 (t, 1 H), 4.66-
4.50 (q, 2 H), 1.55-1.40 (t, 3 H).
Step 2:
[0067] To a suspension of 5-1 (18.84 g, 85.60 mmol) in methanol (anhydrous, 40 mL) at 00C was added allyl amine (5.38 g, 94.2 mmol), with gentle swirling, over a period of 5 min to give a homogeneous solution. The reaction flask was stoppered and allowed to stand at 5 0C for 3 d. The mixture was concentrated in vacuo to give the crude product as viscous yellow oil. The residue was partitioned between 1 N NaOH (100 mL) and DCM (4 x 100 mL). The combined DCM extracts were washed with brine (100 mL), dried over Na2SO4 and concentrated to give 15.62 g (80.1 mmol, 85%) 5-2 as pale yellow oil. Data for 5-2: 1H NMR (CDCl3): δ 7.61 (br m, 1 H), 7.48 (br m, 1 H), 7.40-7.31 (m, 2 H), 6.00 (ddt, 1 H), 5.30 (d, 1 H), 5.21 (d, 1 H), 3.98 (d, 2 H); MS (ESI), m/z (relative intensity, assignment): 195 [100, (M+H)+].
Step 3:
[0068] To a solution of 5-2 (15.6 g, 80.1 mmol) in EtOH (anhydrous, 20 mL) was added diethyl ethoxymethylenemalonate (14.6 mL, 72.9 mmol), and the resultant mixture was heated at reflux for 16 h. The mixture was concentrated in vacuo, and the residue was taken up in ethyl acetate (225 mL) and washed with sat. NH4Cl (2 x 100 mL), H2O (2 x 50 mL) and brine (50 mL). The combined aqueous layers were back-extracted with ethyl acetate (50 mL), and the combined organics were dried over Na2SO4 and concentrated to give 23.39 g (73.4 mmol, 92%) 5-3 as a red-orange, viscous oil.
Data for 5-3: 1H NMR (CDCl3): δ 8.68 (s, 1 H), 7.56-7.52 (m, 2 H), 7.48-7.39 (m, 2 H), 5.93 (ddt, 1 H), 5.27 (d, 1 H), 5.00 (d, 1 H), 4.58 (d, 2 H), 4.41 (q, 2 H), 1.40 (t, 3 H); MS (ESI)5 m/z (relative intensity, assignment): 319 + 321 (100 + 33, [M+H]+), 658 + 660 (12 + 6, PM+Na]*).
Step 4:
[0069] To a solution of 5-3 (512 mg, 1.61 mmol) in pyridine (anhydrous, 2.3 mL) in a 5-dram vial was added lithium iodide (547 mg, 4.09 mmol). The vial was capped and the mixture was heated at 115 0C for 7.5 h. The resultant dark mixture was concentrated in vacuo, the residue was treated with 1 N HCl (5 mL) and the resultant suspension was extracted with 20% MeOH/DCM (3 x 5 mL). The combined organic extracts were washed with 6 N HCl (5 mL), dried over Na2SO4 and concentrated to give a dark brown tar. Trituration with ether gave 361 mg (1.24 mmol, 77%) of 5-4 as brown solid.
Data for 5-4: 1H NMR (/-DMSO): δ 13.19 (br s, 1 H), 8.67 (s, 1 H),7.69-7.64
(m, 2 H), 7.58 (m, 2 H), 5.83 (ddt, 1 H), 5.15 (d, 1 H), 4.92 (d, 1 H), 4.48 (d, 2
H).
Step 5:
[0070] A solution of 5-4 (252 mg, 0.867 mmol) in THF (anhydrous, 5 niL) was cooled to 0 0C and N-methyl morpholine (105 μL, 0.954 mmol) was added, followed by isobutyl chloroformate (112 μL, 0.867 mmol). The mixture was stirred 2 min and N-aminopropylpiperidine (136 mg, 0.954 mmol) was added. The cooling bath was removed, and the mixture was stirred 2 h at rt. The reaction mixture was concentrated in vacuo, and the residue was partitioned between DCM (30 mL) and sat. NaHCO3 (30 mL). The aqueous layer was extracted with DCM (2 x 10 mL) and the combined organics were washed with brine (15 mL), dried over Na2SO4 and concentrated to give crude product as viscous yellow oil. Flash chromatography (5% MeOH/DCM with 0.5% NH4OH) afforded 275 mg (0.663 mmol, 76%) crude 5-5 as a viscous yellow oil. Data for 5-5: MS (ESI), m/z (relative intensity, assignment): 415 + 417 (100 + 30, [M+H]+).
Step 6:
[0071] Compound 5-5 was converted to the corresponding TFA salt by treatment with trifluoroacetic acid (5 mL). The salt was dissolved in 25% MeOH/DCM (10 mL) and the resultant solution was cooled to -78 0C. Ozone was passed through the reaction mixture (ca. 10 min) until a blue color persisted. Oxygen was bubbled through until the blue color of ozone had faded and methyl sulfide (0.24 mL, 3.32 mmol) was added. The mixture was stirred 15 h, then washed with 1 N NaOH (5 mL). The aqueous wash was back- extracted with DCM, and the combined organic extracts were washed with brine, dried over Na2SO4 and concentrated to give 250 mg (0.600 mmol, 90%) crude 5-6 as viscous red oil.
Data for 5-6: MS (ESI), m/z (relative intensity, assignment): 417 + 419 (21 +
16, [M+H]+), 449 + 451 (100 + 30, [M+MeOH]+).
Step 7:
[0072] To a solution of 5-6 in 'BuOH-H2O (4:1, 1 0 mL) was added NaH2PO4 (252 mg, 2.10 mmol), followed by 2-methyl-2-butene (2.0 M solution in THF, 1.8 mL, 3.6 mmol) and sodium chlorite (71 mg,. 0.78 mmol). The resultant mixture was stirred 19 h at rt, then concentrated in vacuo. The residue was taken up in 2 N HCl-MeCN (5 mL) and purified by ion exchange chromatography, eluting with 20% MeCN/H2O until neutral, then H2O-MeCN-NH4OH (4:1:0.5) to provide 204 mg (0.471 mmol, 79%) 5-7 as a white solid. Data for 5-7: 1H NMR (CDCl3): δ 9.51 (br t, 1 H, amide NH), 8.96 (s, 1 H), 7.77 (dd, 1 H), 7.59 (ddd, 1 H), 7.49 (ddd, 1 H), 7.44-7.37 (m, 1 H), 4.49 (br s, 2 H), 3.45-3.43 (br m, 4 H), 2.97-2.92 (br m, 4 H), 2.14-2.05 (br m, 2 H), 1.91 (br m, 6 H); MS (ESI), m/z (relative intensity, assignment): 433 + 435 (100 + 36, [M+H]+).
Step 8:
[0073] To a solution of 5-7 (25.3 mg, 0.0584 mmol) in THF (anhydrous, 0.50 mL) was added N-methylmorpholine (7.7 μL, 0.070 mmol) and isobutyl chloroformate (7.6 μL, 0.058 mmol). The mixture was stirred 2 min and tert- butylamine (7.3 μL, 0.070 mmol) was added. Stirring was continued for 3 d, and the mixture was concentrated in vacuo. The residue was purified by reversed phase preparative HPLC to provide 9.3 mg (0.016 mmol, 27%) 5-8 as the corresponding TFA salt (white solid).
Data for 5-8 (TFA salt): 1H ΝMR (/-MeOH): δ 9.55 (br t, 1 H, amide NH), 8.86 (s, 1 H), 7.85 (br s, 1 H, amide NH), 7.66-7.63 (m, 2 H), 7.59-7.51 (m, 2 H), 4.62 (s, 2 H), 3.58-3.52 (m, 4 H), 3.19-3.14 (t, 2 H), 2.99-2.91 (td, 2 H), 2.12-1.96 (m, 4 H), 1.85-1.72 (m, 3 H), 1.61-1.50 (m, 1 H), 1.31 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 489 + 491 (100 + 46, [M+H]+), 999 + 1001 (5 + 2, [2M+Na]+).
Example 6. Preparation of 6-8.
Figure imgf000044_0001
NaCIO2, 2-Me-2-butene 1BuNH2 KH2PO4, 1BUOH-H2O (4:1) EDCl, HOBt, DMF DCM
Figure imgf000044_0004
45% for 3 steps
Figure imgf000044_0002
CI 100%
Figure imgf000044_0003
6-4 6-5 6-6
Figure imgf000044_0005
Step 1:
[0074] Compound 5-4 (820 mg, 2.82 mmol) was dried via azeotropic removal of water with toluene (2 x 35 mL), then taken up in tert-butanol (anhydrous, 8.5 niL). Triethylamine (0.39 mL, 2.8 mmol) and diphenylphosphoryl azide (0.61 mL, 2.8 mmol) were added, and the reaction mixture was heated at reflux for 28 h. The mixture was concentrated in vacuo and the residue was taken up in ethyl acetate (40 mL), washed with sat. NaHCO3 (2 x 40 mL),H20 (40 mL) and brine (40 mL). The organic layer was dried over sodium sulfate and concentrated in vacuo to give the crude product as yellow-brown oil. Flash chromatography (10% ethyl acetate/hexanes) gave 379 mg (1.05 mmol, 37%) X-8 as white solid. Data for 6-1: 1H NMR (CDCl3): δ 8.68 (br s, 1 H), 7.49-7.46 (m, 2 H), 7.42- 7.34 (m, 3 H), 5.87 (ddt, 1 H), 5.24 (d, 1 H), 4.95 (d, 1 H), 4.55 (d, 2 H), 1.53 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 306 + 308 (100 + 32, [MH - 56]+), 362 + 364 (58 + 13, [M +H]+).
Step 2:
[0075] To a stirred solution of 6-1 (253 mg, 0.699 mmol) in THF (6 mL) was added a solution of N-methylmorpholine oxide (119 mg, 1.02 mmol) in H2O (1 mL), followed by a 2.5% solution (w/w, 263 μL, 0.021 mmol, 3 mol%) of osmium tetroxide in fert-butanol. The resultant mixture was stirred 20 h at rt. Saturated Na2S2O3 (aq., 1.5 mL) and Celite were added, and stirring was maintained for 30 min. The suspension was filtered, and the filter cake was washed with ethyl acetate (3 x 10 mL). The combined filtrates were concentrated and the residue was redissolved in 25% IPA/DCM (15 mL). The resultant solution was washed with sat. Na2S2O3 (10 mL), 10% NaHSO4 (10 mL) and brine (10 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo to give 227 mg (0.573 mmol, 82%) 6-2 as white foam. Data for 6-2: MS (ESI), m/z (relative intensity, assignment): 340 + 342 (100, [MH - 56]+), 396 + 398 (23 + 9, [M+H]+), 418 + 420 [23 + 6, [M+Naf).
Step 3:
[0076] To a stirred solution of 6-2 (227 mg, 0.573 mmol) in abs. EtOH (2 mL) was added a solution of sodium periodate (245 mg, 1.15 mmol) in H2O (0.60 mL). The resultant milky suspension was stirred 1.5 h at rt, then filtered through Celite. The filter cake was washed with 25% IPA/DCM (3 x 10 mL), and the combined filtrates were concentrated in vacuo to give 302 mg (sample + residual IPA) of crude 6-3, which was immediately used in the next step without further purification. Data for 6-3: 1H NMR (CDCl3) δ 9.62 (s, 1 H)5 8.73 (br s, 1 H), 7.50-7.37 (m, 5 H), 4.78 (s, 2 H), 1.53 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 340 + 342 (55 + 14, [MH - 56]+), 396 + 398 (100 + 35, [M + MeOH]+).
Step 4:
[0077] To a stirred solution of 6-3 in tert-butanόi (9 mL) was added a solution of sodium hydrogen phosphate (202 mg, 1.68 mmol) in H2O (2 mL). To this were added NaClO2 (1.3 eq) and 2-methyl-2-butene (6.0 eq). The resultant solution was stirred 20 h at rt, then concentrated in vacuo to give a white solid. The residue was partitioned between H2O (15 mL) and 25% MeOH/DCM (20 mL). The aqueous layer was extracted with 25% MeOH/DCM (2 x 20 mL) and the combined organic extracts were washed with brine (15 mL), dried over Na2SO4 and concentrated in vacuo to give 184 mg crude 6-4 as white solid. Data for 6-4: 1H NMR (CDCl3) δ 8.71 (br s, 1 H), 7.50-7.34 (M, 5 H), 4.60 (s, 2 H), 1.53 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 324 + 326 (51 + 24, [MH - 56]+), 380 + 382 (100 + 26, [MH-H]+).
Step 5:
[0078] To a stirred solution of 6-4 in DMF (anhydrous, 2.5 mL) was added tert- butylamine (56 μL, 0.53 mmol), followed by EDCI (184 mg, 0.96 mmol) and HOBt hydrate (195 mg, 1.4 mmol). The reaction mixture was stirred 16 h at rt, then partitioned between sat. NaHCO3 (20 mL) and ethyl acetate (30 mL). The aqueous phase was extracted with ethyl acetate (2 x 25 mL), and the combined organic extracts were washed with brine (20 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by flash chromatography (25% ethyl acetate/hexanes) to give 93.9 mg (0.216 mmol, 45% from 6-2) 6-5 as white foam.
Data for 6-5: 1H NMR (CDCl3) δ 8.69 (br s, 1 H), 7.57 (dd, 1 H), 7.49-7.36 (m, 3 H), 7.29 (s, 1 H), 5.53 (br s, 1 H), 4.42 (s, 2 H), 1.52 (s, 9 H), 1.35 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 379 + 381 (22 + 5, [MH - 56]+, 435 + 437 (100 + 45, [M+H]+).
Step 6:
[0079] Trifluoroacetic acid (1.5 niL) was added to a solution of 6-5 (93.9 mg,
0.216 mmol) in DCM (anhydrous, 1.5 mL). The resultant yellow solution was stirred 40 min at rt and concentrated in vacuo to give 125 mg crude 6-6 as yellow-brown foam.
Data for 6-6: MS (ESI), m/z (relative intensity, assignment): 335 + 337 (100,
[M+H]+).
Step 7:
[0080] To a stirred solution of crude 6-6 (19.9 mg, 0.046 mmol) in THF (anhydrous, 0.5 mL) at 0 0C was added sodium carbonate (22 mg, 0.21 mmol) and 4-chlorobutyryl chloride (7.0 μL, 0.060 mmol). The reaction mixture was stirred 1 h at 0 0C, poured into sat. NaHCO3 (1 mL) and extracted with ethyl acetate (2 x 3 mL). The combined organic extracts were washed with brine (2 mL), dried over Na2SO4 and concentrated in vacuo to give 11.5 mg (0.026 mmol, 76%) 6-7 as off-white foam.
Data for 6-7: 1H NMR (CDCl3) δ 9.07 (s, 1 H), 8.04 (br s, 1 H), 7.58 (t, 1 H), 7.58-7.37 (m, 3 H), 5.48 (br s, 1 H), 4.44 (s, 2 H), 3.66 (t, 2 H), 2.64 (t, 2 H), 2.20 (app quint, 2 H), 1.36 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 439 + 441 (100 + 58, [M+H]+), 461 + 463 (26 + 19, [M+Na]+).
Step 8:
[0081] To a stirred solution of 6-7 (11.5 mg, 0.026 mmol) in MeCN (anhydrous, 0.3 mL) was added K2CO3 (18 mg, 0.13 mmol), piperidine (7.0 μL, 0.080 mmol) and a catalytic amount of sodium iodide. The resultant mixture was heated 8 h at 85 0C, then poured into ethyl acetate (4 mL). The layers were separated, and the aqueous phase was extracted with ethyl acetate (3 x 1 mL). The combined organic extracts were washed with brine (1 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by preparative TLC (5% MeOH/DCM + 0.5% NH4OH, 500 μm) to give 2.5 mg (0.0051 mmol, 20%) 6-8 as a pale yellow glassy solid.
Data for 6-8: 1H NMR (CDCl3) δ 9.07 (s, 1 H), 8.28 (br s, 1 H), 7.58 (t, 1 H), 7.50-7.45 (m, 3 H), 5.56 (br s, 1 H), 4.44 (s, 2 H), 2.55-2.50 (m, 4 H), 2.04-1.96 (m, 4 H), 1.75-1.66 (br m, 4 H), 1.51-1.40 (br m, 4 H), 1.36 (s, 9 H); MS (ESI), m/z (relative intensity, assignment): 488 + 490 (100 + 34, [M+H]+).
Example 7. Preparation of 7-4.
Figure imgf000048_0001
Step 1:
[0082] Compound 7-1 was prepared from literature procedures (J. Med. Chem., 2003, 46, 4050-4062). To a solution of compound 7-1 (1.0 g, 2.4 mmol) in acetonitrile (25 mL) was added l-(2-(pyrrolidin-3-yl)ethyl)piperidine trifluoroacetic acid (666 mg, 2.4 mmol) and potassium carbonate (660 mg, 2.4 mmol). The mixture was stirred at reflux for 16 h. After cooling to rt, the mixture was filtered and the filtrate was concentrated. The filtrate was then diluted with CH2Cl2 (3OmL) and washed with saturated brine solution. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by flash chromatography to afford 7-2 (770 mg, 1.36 mmol, 57%). Data for 7-2: MS (ESI), m/z (assignment): 565.3/567.3 ([M+H]+, 100/35).
Step 2:
[0083] To a solution of compound 7-2 (385 mg, 0.68 mmol) in methanol (2 mL) was added catalytic amount of 10% Pd/C. The reaction mixture was stirred at 50 0C under 50 psi of hydrogen for 16 h. After cooling to rt, the reaction was filtered and filtrate was evaporated to afford compound 7-3 (300 mg, 0.68 mmol, quantitative).
Data for 7-3: MS (ESI), m/z (assignment): 441.2 ([M+H]+, 100).
Step 3:
[0084] To a solution of compound 7-3 (100 mg, 0.23 mmol) in acetonitrile (5 mL) was added propan-2-amine (100 uL, 5 equiv.), EDCI (1.15 mmol) and HOBt (1.15 mmol). The reaction was stirred at reflux for 2 h. After cooling to rt, the mixture was filtered and the filtrate was concentrated. The filtrate was then diluted with CH2Cl2 (5 mL) and washed with saturated NaHCO3 and brine. The organic layer was dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC to afford 7-4 (30 mg, 0.062 mmol, 27%).
Data for 7-4: 1H NMR (400 MHz, CDCl3) δ; 7.87 (br s, IH), 7.30 (t, IH), 6.90-6.99 (s+d, 3H), 6.63 (s, IH), 4.29 (s, 2H), 3.99 (septet, IH), 3.85-3.77 (m+s, 7H), 3.55 (br m, 2H), 3.09 (br m, 2H), 2.69 (br m, 2H), 2.34-2.23 (m, 2H), 1.50-2.20 (m, 8H), 1.42 (br s, IH), 1.08 (d, 6H); MS (ESI), m/z (assignment): 482.2 ([M+H]+, 100). Example 8. Preparation of 8-3.
Figure imgf000050_0001
Figure imgf000050_0002
[0085] Compound 8-1 was prepared from similar procedures in Example 7. To a solution of 8-1 (90 mg, 0.17 mmol) in methanol (2 mL) was added 10% Pd/C (catalytic amount). The reaction was stirred under 1 arm of hydrogen for 16 h. The mixture was filtered and evaporated to afford 8-2 (70 mg, 0.16 mmol, 94%). Compound 8-2 was coupled with isopropylamine by using the same procedures in Example 7 to give 8-3. Data for 8-3: MS (ESI), m/z (assignment): 490/492 ([M+H]+, 100/35).
[0086] Scheme for pyridone, IV:
Figure imgf000050_0003
Analogous to description
Figure imgf000051_0001
for pyridone in Example 3.
Figure imgf000051_0002
Synthesis described in Lin et a/ J. Org. Chem. 2003, 68, 5688
Figure imgf000051_0003
Other compounds may be prepared by modifying the synthesis of starting
material A, for example to prepare
Figure imgf000051_0004
as the starting material. The article of Lin et al., J. Org. Chem., 2003, 68:5688, is incorporated herein by reference.
[0087] Scheme for pyridazin-3-one VI:
Figure imgf000051_0005
Figure imgf000052_0001
Condensation
Figure imgf000052_0002
Figure imgf000052_0003
Analogous to description for pyridone synthesis
Figure imgf000052_0004
[0088] Scheme for phenyl VII:
Figure imgf000052_0005
Figure imgf000052_0006
Figure imgf000053_0001
Bromination in first step seeJACS, 123(15), 3434-3440; 2001
U.S. Patent No. 6307047 and the article from J. Am. Chem. Soc, 123(15):3434- 3440 (2001) are incorporated herein by reference.
[0089] Scheme for pyridine VEQ.:
heteroannulation see: Org. Lett. 2000, 2, 2339-41.
Figure imgf000053_0002
Lett.
Figure imgf000053_0003
Figure imgf000053_0004
analogous to pyridone synthesis
Figure imgf000054_0001
The articles from Org. Lett. 2000, 2, 2339-41 and Org. Lett. 6(17)2837-2840 are incorporated herein by reference.
[0090] Scheme for pyrimidine IX:
Figure imgf000054_0002
Figure imgf000055_0001
[0091] Scheme for pyrazine X:
Figure imgf000055_0002
Compound A reported in Miesel et al, US 4,160,834 Transformation analogous to A to B and C to D reported in Thompson et al, J. Org. Chem. 1988, 53, 2052-2055
U.S. Pat. No. 4,160,834 and the article of Thompson et al., J. Org. Chem. 1988, 2052-2055 are incorporated herein by reference.
Assay:
[0092] Chinese Hamster Ovary (CHO) cells stably expressing the human V3 receptor were incubated to equilibrium with the test compound (at a final assay concentration of 10"1VoLL"1 to 10"VoLL"1) and [3H]AVP (at a final assay concentration of 5 x 10"9 mol.L"1). Throughout the concentration of dimethylsulphoxide (DMSO) did not exceed 1% (v/v). After washing with room temperature phosphate buffered saline (PBS), scintillation fluid was added and the plates counted on a MicroBeta Trilux counter. A sigmoidal dose response curve (non-linear regression, variable slope) was plotted as concentration of test compound (mol.L"1) against percentage specific binding of [3H]AVP and an IC50 value was calculated.
[0093] Table 1 shows compounds that exhibited IC50 less than 10 μM:
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000057_0003
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000058_0003
Figure imgf000059_0001
Figure imgf000059_0002
Figure imgf000059_0003

Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000060_0003
Figure imgf000061_0001
Figure imgf000061_0002
Figure imgf000061_0003
Figure imgf000062_0001
Figure imgf000062_0002

Claims

CLAIMSWe claim:
1. A compound of formula:
Figure imgf000063_0001
wherein
X4-X5 is chosen from CR4-CR5, N-CR5 and CR4-N; one of X6, X7 and X8 is N and the other two are CR4 and CR5;
R1 is chosen from (Ci.io)alkyl, (C3-1O)CyClOaIlCyI, and [(C3-1O)CyClOaIlCyI(C1- 2)alkyl], said (C1-1o)alkyl, (C3-1 o)cycloalkyl, and [(C3-Io)CyClOaIlCyI(C1- 2)alkyl] heing optionally substituted with one or more halogens, (C1- 6)alkoxy, (C2-6)alkenyl, (C2-6)alkynyl, phenyl or benzyl; Ar is chosen from
(i) (C6-io)aryl, optionally substituted within 1-3 substituents selected from halogen, hydroxy, cyano, COOR5, NR6R7, phenyl, (C5-6)heteroaiyl, (C1-
6)alkyl, (C3-δ)cycloalkyl, (Ci-6)alkyloxy and (C3-6)cycloalkyloxy, said (C1- δ)alkyl, (C3-6)cycloalkyl, (Ci-6)alkyloxy and (C3.6)cycloalkyloxy being optionally substituted with one or more halogens;
(ii) (C5-io)heteroaryl optionally substituted with a substituent selected from methyl, (C1-6)alkyloxy or halogen; and
(iii) (C4-7)cycloallcyl; R4 and R5 are independently chosen from H, (C^alkyl, (C1-6)alkyloxy or halogen, said (C1-6)alkyl, (C1-6)alkyloxy being optionally substituted with one or more halogens;
G is a linking moiety spanning 4 to 7 atoms between termini; and R8 and R9 are residues that, in combination, maintain the basicity of N, with the proviso that when X4-X5 is CR4-N and G is alkylene, R1 is chosen from (C1-6)alkyl, (C3-6)cycloalkyl and [(C3-6)cycloalkyl (C1-2)alkyl].
2. A compound according to claim 1 wherein G is a linking moiety spanning 4 to 7 atoms between termini, said linking moiety incorporating at least one of
(a) an sp2 hybridized carbon,
(b) a cyclic structure, and
(c) when the nitrogen bearing R8 and R9 is part of a 4- to 7-membered nitrogenous heterocycle, G may additionally be -(C4-7)alkylene or -N(R10)-(C3- 6)alkylene; and R , 1i0υis H or (C1-6)alkyl.
3. A compound according to claim 1 wherein at least one of R8 and R9 is other than hydrogen.
4. A compound according to claim 3 of formula
Figure imgf000065_0001
wherein
R1 is C3-C6 alkyl;
R10 is H or (C1-6)alkyl; and
Figure imgf000066_0001
is a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings.
5. A compound according to claim 1 wherein both of R8 and R9 are other than hydrogen.
6. A compound according to claim 1 of formula:
Figure imgf000066_0002
wherein X4-X5 is chosen from CR4-CR5, N-CR5 and CR4-N;
R1 is chosen from (Ci.io)alkyl, (C3,io)cycloalkyl,
Figure imgf000066_0003
said (Ci-Io)alkyl, (C3.io)cycloalkyl, and [(C3-io)cycloalkyl(C1-2)alkyl] being optionally substituted with one or more halogens, (Q^alkoxy, (C2- 6)alkenyl, (C2-6)alkynyl, phenyl or benzyl;
Ar is (C6-1o)aryl, optionally substituted within 1-3 substituents selected from halogen, hydroxy, cyano, COOR5, NR6R7, phenyl, (C5-6)heteroaryl, (Ci- 6)alkyl, (C3-6)cycloalkyl, (Ci-6)alkyloxy and (C3-6)cycloalkyloxy, said (Ci- 6)alkyl, (C3-6)cycloalkyl, (Ci^alkyloxy and (Cs-^cycloalkyloxy being optionally substituted with one or more halogens, or Ar is a (C5-io)heteroaryl optionally substituted with a substituent selected from methyl, (Ci- 6)alkyloxy or halogen or R2 is (C4-7)cycloalkyl; R4 and R5 are independently chosen from H, (C^alkyl, (C1-6)alkyloxy or halogen, said (C1-6)alkyl, (Ci-6)alkyloxy being optionally substituted with one or more halogens;
G is a linking moiety spanning 4 to 7 atoms between termini, said linking moiety incorporating at least one of
(a) an sp2 hybridized carbon,
(b) a cyclic structure, and
(c) when the nitrogen bearing R8 and R9 is part of a 4- to 7-membered nitrogenous heterocycle, G may additionally be -(C4-7)alkylene or -N(R10)-(C3- 6)alkylene;
R8 and R9 are non-hydrogen residues that, in combination, maintain the basicity of N; and
R , 1l0uis H or (Ci-6)alkyl.
7. A compound according to claim 6 of formula:
Figure imgf000067_0001
A compound according to claim 6 of formula:
Figure imgf000068_0001
9. A compound according to claim 6 of formula:
Figure imgf000068_0002
10. A compound according to claim 6 chosen from formulae:
Figure imgf000068_0003
Figure imgf000069_0001
wherein
E is (C2-1o)hydrocarbon; and
Figure imgf000069_0002
is a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings.
11. A compound according to claim 10 wherein
Figure imgf000069_0003
is a piperidine or morpholine ring.
12. A compound according to claim 6 of formula:
Figure imgf000069_0004
wherein J is a direct bond or (C^hydrocarbon; and
Figure imgf000070_0001
is a single ring carbocycle or heterocycle of 4 to 7 atoms or a two ring carbocycle or heterocycle of 9 to 13 atoms.
13. A compound according to claim 12 wherein
Figure imgf000070_0002
is a five or six- membered nitrogenous heterocycle and J is methylene, ethylene or propylene.
14. A compound according to claim 13 wherein -NR 8r R>9 . is chosen from
Figure imgf000070_0003
and -N[C1-3alkyl]2
15. A compound according to claim 6 of formula:
Figure imgf000070_0004
of 6 to 8 atoms or a two ring heterocycle of 9 to 13 atoms in which the nitrogen labeled b is the nitrogen of claim 1 and the nitrogen labeled a is subsumed in the definition of G in claim 1.
16. A compound according to claim 15 wherein
Figure imgf000071_0001
is a hexahydro-l,4-diazepine ring.
17. A compound according to claim 6 chosen from formulae:
Figure imgf000071_0002
wherein
Figure imgf000072_0001
a saturated nitrogenous heterocycle of 3 to 10 carbons in one or two rings; and R8 is chosen from hydrogen and C1-I0 hydrocarbon.
18. A compound according to claim 17 wherein
Figure imgf000072_0002
is a piperidine ring and R8 is methyl.
19. A compound chosen from formulae:
Figure imgf000072_0003
wherein X4-X5 is chosen from CR4-CR5, N-CR5 and CR4-N; R1 is chosen from (C1-1o)alkyl, (C3-6)cycloalkyl, (C3-6)cycloalkyl (C1-2)alkyl, said (C1-Io)alkyl, (C3-6)cycloalkyl, and (C3-6)cycloalkyl (C1-2)alkyl being optionally substituted with one or more halogens, (C2-δ)alkenyl, (C2- 6)alkynyl, phenyl or benzyl;
Ar is
Figure imgf000073_0001
optionally substituted within 1-3 substituents selected from halogen, hydroxy, cyano, COOR5, NR6R7, phenyl, (C5-6)heteroaryl, (Ci- 6)alkyl, (C3-6)cycloalkyl, (Ci.6)alkyloxy and (C3-6)cycloalkyloxy, said (Ci- 6)alkyl, (C3-6)cycloalkyl, (Ci^alkyloxy and (C3-6)cycloalkyloxy being optionally substituted with one or more halogens, or Ar is a (C5-io)heteroaryl optionally substituted with a substituent selected from methyl, (Ci- 6)alkyloxy or halogen or R2 is (C4-7)cycloalkyl;
R4 and R5 are independently chosen from H, (
Figure imgf000073_0002
Ci^alkyl, or halogen, said (C^alkyl, (Ci-6)alkyloxy being optionally substituted with one or more halogens;
R10 is H or (Ci-6)alkyl; and
Pyr is chosen from imidazole and optionally substituted pyridine attached through a carbon.
20. A compound according to any of claims 1-3 and 5-19 wherein R1 is chosen from C4-6alkyl, cycloalkyl and Ci-3alkyl substituted with phenyl, methoxy or alkynyl.
21. A compound according to claim 20 wherein R1 is chosen from t-butyl, isopropyl, cyclopentyl, α-methylbenzyl, methoxypropyl and propargyl.
22. A compound according to any of claims 1-19 wherein Ar is chosen from phenyl and phenyl substituted with halogen, methyl or methoxy.
23. A pharmaceutical formulation comprising a pharmaceutically acceptable carrier and a compound according to any of claims 1-19.
24. A method for effecting diuresis or treating a disorder chosen from depression, stress disorders, aggressive behavior, insomnia in elderly patients, cancer, Cushing's Disease, and pancreatic disease, comprising administering a compound according to any of claims 1-19.
25. The use of a compound according to any one of claims 1-19 for the manufacture of a medicament.
26. Use according to claim 25 wherein said medicament is for effecting diuresis or for treating a disorder chosen from depression, stress disorders, aggressive behavior, insomnia in elderly patients, cancer, Cushing's Disease, and pancreatic disease.
PCT/US2006/022025 2005-06-07 2006-06-07 Azinone and diazinone v3 inhibitors for depression and stress disorders WO2006133242A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU2006255009A AU2006255009B2 (en) 2005-06-07 2006-06-07 Azinone and diazinone V3 inhibitors for depression and stress disorders
CA002610400A CA2610400A1 (en) 2005-06-07 2006-06-07 Azinone and diazinone v3 inhibitors for depression and stress disorders
MX2007015427A MX2007015427A (en) 2005-06-07 2006-06-07 Azinone and diazinone v3 inhibitors for depression and stress disorders.
EP06760716.8A EP1890697B1 (en) 2005-06-07 2006-06-07 Azinone and diazinone v3 inhibitors for depression and stress disorders
BRPI0611348-6A BRPI0611348A2 (en) 2005-06-07 2006-06-07 compound, pharmaceutical formulation, method for diuresis or treatment of a disorder, and use of a compound
CN2006800237203A CN101212969B (en) 2005-06-07 2006-06-07 Azinone and diazinone V3 inhibitors for depression and stress disorders
JP2008515847A JP5002592B2 (en) 2005-06-07 2006-06-07 Azinone and diazinon V3 inhibitors for depression and stress disorders
IL187634A IL187634A0 (en) 2005-06-07 2007-11-26 Azinone and diazinone v3 inhibitors for depression and stress disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68830605P 2005-06-07 2005-06-07
US60/688,306 2005-06-07

Publications (2)

Publication Number Publication Date
WO2006133242A2 true WO2006133242A2 (en) 2006-12-14
WO2006133242A3 WO2006133242A3 (en) 2007-02-15

Family

ID=37309794

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/022025 WO2006133242A2 (en) 2005-06-07 2006-06-07 Azinone and diazinone v3 inhibitors for depression and stress disorders

Country Status (10)

Country Link
US (1) US8350043B2 (en)
EP (1) EP1890697B1 (en)
JP (1) JP5002592B2 (en)
CN (1) CN101212969B (en)
AU (1) AU2006255009B2 (en)
BR (1) BRPI0611348A2 (en)
CA (1) CA2610400A1 (en)
IL (1) IL187634A0 (en)
MX (1) MX2007015427A (en)
WO (1) WO2006133242A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011031658A1 (en) 2009-09-09 2011-03-17 E. I. Du Pont De Nemours And Company Herbicidal pyrimidone derivatives
WO2012033548A2 (en) 2010-09-07 2012-03-15 E. I. Du Pont De Nemours And Company Herbicidal bis-nitrogen-containing oxo and sulfono heterocycles
WO2012043791A1 (en) 2010-10-01 2012-04-05 大正製薬株式会社 1,2,4-triazolone derivative
WO2012122171A1 (en) 2011-03-09 2012-09-13 E. I. Du Pont De Nemours And Company Herbicidal bis-nitrogen-containing oxo heterocycles
WO2012148622A1 (en) 2011-04-28 2012-11-01 E. I. Du Pont De Nemours And Company Herbicidal pyrazinones
WO2013062027A1 (en) 2011-10-27 2013-05-02 大正製薬株式会社 Azole derivative
WO2013147117A1 (en) 2012-03-30 2013-10-03 大正製薬株式会社 Fused azole derivative
US9604965B2 (en) 2010-04-23 2017-03-28 Cytokinetics, Inc. Substituted pyridazines as skeletal muscle modulators
US9730886B2 (en) 2010-04-23 2017-08-15 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US9994528B2 (en) 2010-04-23 2018-06-12 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL1928454T3 (en) 2005-05-10 2015-03-31 Intermune Inc Pyridone derivatives for modulating stress-activated protein kinase system
US8304413B2 (en) 2008-06-03 2012-11-06 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
CN102731414B (en) * 2012-07-04 2014-11-26 宁波九胜创新医药科技有限公司 Method for preparing 4-[(4-chloro-2-pyrimidinyl) amino] cyanophenyl
AR092742A1 (en) 2012-10-02 2015-04-29 Intermune Inc ANTIFIBROTIC PYRIDINONES
WO2014160592A2 (en) * 2013-03-27 2014-10-02 Merck Sharp & Dohme Corp. FACTOR XIa INHIBITORS
JP6387669B2 (en) * 2013-04-26 2018-09-12 大正製薬株式会社 Medicaments containing azole derivatives
MX2016012808A (en) 2014-04-02 2017-01-05 Intermune Inc Anti-fibrotic pyridinones.
AU2019239658A1 (en) * 2018-03-21 2020-11-12 Suzhou Puhe BioPharma Co., Ltd. SHP2 inhibitors and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055130A2 (en) 2000-01-25 2001-08-02 Sanofi-Synthelabo Novel 1,3-dihydro-2h-indol-2-one derivatives and their use as ligands for v1b and v1a arginine-vasopressin receptors
WO2004009585A2 (en) 2002-07-19 2004-01-29 Sanofi-Aventis Acyloxypyrrolidine derivatives and use thereof as ligands of v1b or both v1b and v1a receptors

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521179A (en) * 1991-04-18 1996-05-28 Zeneca Limited Heterocyclic amides
GB9207145D0 (en) * 1991-04-18 1992-05-13 Ici Plc Heterocyclic amides
CA2263999A1 (en) * 1996-08-29 1998-03-05 Merck & Co., Inc. Integrin antagonists
JP2002179651A (en) * 1998-06-19 2002-06-26 Wakamoto Pharmaceut Co Ltd Benzanilide derivative and medicine composition
MXPA01011804A (en) * 1999-05-19 2003-09-04 Pharmacia Corp Substituted polycyclic aryl and heteroaryl pyrymidinones useful as anticoagulants.
US6664255B1 (en) 1999-05-19 2003-12-16 Pharmacia Corporation Substituted polycyclic aryl and heteroaryl pyrazinones useful for selective inhibition of the coagulation cascade
US6908919B2 (en) 1999-05-19 2005-06-21 Pharmacia Corporation Substituted polycyclic aryl and heteroaryl pyrazinones useful for selective inhibition of the coagulation cascade

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055130A2 (en) 2000-01-25 2001-08-02 Sanofi-Synthelabo Novel 1,3-dihydro-2h-indol-2-one derivatives and their use as ligands for v1b and v1a arginine-vasopressin receptors
WO2004009585A2 (en) 2002-07-19 2004-01-29 Sanofi-Aventis Acyloxypyrrolidine derivatives and use thereof as ligands of v1b or both v1b and v1a receptors

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
BERNARDINI ET AL., NEUROENDOCRINOLOGY, vol. 60, 1994, pages 503 - 508
BLANCHARD ET AL., PHARMCOL. BIOCHEM. BEHAV., vol. 80, 2005, pages 189 - 194
CHEN ET AL., J. NEUROSCI. RES., vol. 60, 2000, pages 761 - 766
DAHIA ET AL., J. CLIN. ENDOCRIN. METAB., vol. 81, 1996, pages 1768 - 1771
DE GOEIJ ET AL., NEUROENDOCRINOLOGY, vol. 53, 1991, pages 150 - 159
FOLNY ET AL., AM. J. PHYSIOL., vol. 285, 2003, pages E566 - 576
FREIDINGER; PETTIBONE, MEDICINAL RESEARCH REVIEWS, vol. 17, 1997, pages 1 - 16
HOLSBOER ET AL., BIOL. PSYCH., vol. 21, 1986, pages 601 - 611
J. MED. CHEM., vol. 46, 2003, pages 4050 - 4062
KALAMATIANOS ET AL., J. NEUROENDOCRINOL., vol. 16, 2004, pages 493 - 501
MAEHR J., CHEM. ED., vol. 62, 1985, pages 114 - 120
PERRAUDIN ET AL., J. CLIN. ENDOCRIN. METAB., vol. 80, 1995, pages 2661 - 2667
PLOTSKY; MEANEY, MOL. BRAIN RES., vol. 18, 1993, pages 195 - 200
SCOTT; DINAN, LIFE SCIENCES, vol. 62, 1998, pages 1985 - 1988
SPERRY; CARLSON: "DSM-IV diagnosis to treatment", 1996, TAYLOR & FRANCIS
SUGIMOTO ET AL., J. BIOL. CHEM., vol. 269, 1994, pages 27088 - 27092
WERSINGER ET AL., HORM. BEHAV., vol. 46, 2004, pages 638 - 645
WERSINGER ET AL., MOL. PSYCHIATRY, vol. 7, 2002, pages 975 - 984

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9630928B2 (en) 2009-09-09 2017-04-25 E. I. Du Pont De Nemours And Company Intermediates to prepare herbicidal pyrimidone derivatives
US9187432B2 (en) 2009-09-09 2015-11-17 E I Du Pont De Nemours And Company Intermediates to prepare herbicidal pyrimidone derivatives
WO2011031658A1 (en) 2009-09-09 2011-03-17 E. I. Du Pont De Nemours And Company Herbicidal pyrimidone derivatives
AU2010292400B2 (en) * 2009-09-09 2016-09-08 Fmc Corporation Herbicidal pyrimidone derivatives
US8952024B2 (en) 2009-09-09 2015-02-10 E I Du Pont De Nemours And Company Herbicidal pyrimidone derivatives
US10765624B2 (en) 2010-04-23 2020-09-08 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US9994528B2 (en) 2010-04-23 2018-06-12 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use
US9730886B2 (en) 2010-04-23 2017-08-15 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US10076519B2 (en) 2010-04-23 2018-09-18 Cytokinetics, Inc. Substituted pyridazines as skeletal muscle modulators
US10272030B2 (en) 2010-04-23 2019-04-30 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US9604965B2 (en) 2010-04-23 2017-03-28 Cytokinetics, Inc. Substituted pyridazines as skeletal muscle modulators
US11369565B2 (en) 2010-04-23 2022-06-28 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
WO2012033548A3 (en) * 2010-09-07 2015-09-03 E. I. Du Pont De Nemours And Company Herbicidal pyrimidone derivatives
WO2012033548A2 (en) 2010-09-07 2012-03-15 E. I. Du Pont De Nemours And Company Herbicidal bis-nitrogen-containing oxo and sulfono heterocycles
RU2566754C2 (en) * 2010-10-01 2015-10-27 Тайсо Фармасьютикал Ко., Лтд. 1,2,4-triazolone derivative
US9193695B2 (en) 2010-10-01 2015-11-24 Taisho Pharmaceutical Co., Ltd. 1, 2, 4-triazolone derivative and use thereof as an antagonist on the arginine-vasopressin 1B receptor
JP5787237B2 (en) * 2010-10-01 2015-09-30 大正製薬株式会社 1,2,4-triazolone derivatives
AU2011308403B2 (en) * 2010-10-01 2014-08-21 Taisho Pharmaceutical Co., Ltd. 1,2,4-triazolone derivative
KR101820185B1 (en) 2010-10-01 2018-01-18 다이쇼 세이야꾸 가부시끼가이샤 1,2,4-triazolone derivative
WO2012043791A1 (en) 2010-10-01 2012-04-05 大正製薬株式会社 1,2,4-triazolone derivative
WO2012122171A1 (en) 2011-03-09 2012-09-13 E. I. Du Pont De Nemours And Company Herbicidal bis-nitrogen-containing oxo heterocycles
WO2012148622A1 (en) 2011-04-28 2012-11-01 E. I. Du Pont De Nemours And Company Herbicidal pyrazinones
US9522914B2 (en) 2011-10-27 2016-12-20 Taisho Pharmaceutical Co., Ltd Azole derivative
RU2622639C2 (en) * 2011-10-27 2017-06-19 Тайсо Фармасьютикал Ко., Лтд. Azole derivatives
KR20140081824A (en) 2011-10-27 2014-07-01 다이쇼 세이야꾸 가부시끼가이샤 Azole derivative
WO2013062027A1 (en) 2011-10-27 2013-05-02 大正製薬株式会社 Azole derivative
WO2013147117A1 (en) 2012-03-30 2013-10-03 大正製薬株式会社 Fused azole derivative

Also Published As

Publication number Publication date
AU2006255009A1 (en) 2006-12-14
AU2006255009B2 (en) 2011-10-27
EP1890697B1 (en) 2015-03-04
CA2610400A1 (en) 2006-12-14
CN101212969A (en) 2008-07-02
MX2007015427A (en) 2008-04-16
IL187634A0 (en) 2008-03-20
EP1890697A2 (en) 2008-02-27
BRPI0611348A2 (en) 2010-12-07
JP5002592B2 (en) 2012-08-15
JP2008542444A (en) 2008-11-27
US20070037822A1 (en) 2007-02-15
CN101212969B (en) 2013-08-07
US8350043B2 (en) 2013-01-08
WO2006133242A3 (en) 2007-02-15

Similar Documents

Publication Publication Date Title
AU2006255009B2 (en) Azinone and diazinone V3 inhibitors for depression and stress disorders
EP3325481B1 (en) Compounds useful for treating disorders related to kit and pdgfr
CA2583907C (en) Heteroaryl-substituted alkyne compounds and method of use
EP2802577B1 (en) Triazolyl-substituted pyridyl compounds useful as kinase inhibitors
CA2930060C (en) Alkyl-amide-substituted pyridyl compounds useful as modulators of il-12, il-23 and/or ifnalpha responses
AU2017287553B2 (en) Imidazopyrazinamine phenyl derivative and use thereof
EP3990436A1 (en) 2,3-dihydroquinazolin compounds as nav1.8 inhibitors
AU2005300736B2 (en) Anthranilamide pyridinureas as VEGF receptor kinase inhibitors
TW593280B (en) CRF antagonistic quino- and quinazolines
US9321729B2 (en) Substituted pyridyl amide compounds as modulators of the histamine H3 receptor
JP4988583B2 (en) Novel anthranilamidopyridine ureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
EP2463276A1 (en) Nitrogenous-ring acylguanidine derivative
EA029054B1 (en) Urea and amide derivatives of aminoalkylpiperazines and use thereof
WO2018227058A1 (en) Piperidinone formyl peptide 2 receptor agonists
WO2011061318A1 (en) Heterocylic compounds as antagonists of the orexin receptors
KR20120101551A (en) Bicyclic thiazoles as allosteric modulators of mglur5 receptors
US20040259904A1 (en) Fused tri and tetra-cyclic pyrazole kinase inhibitors
CN116601153A (en) Nitrogen-containing 2, 3-dihydroquinazolinone compounds as NAV1.8 inhibitors
EP3634409B1 (en) Orexin receptor antagonists
WO2018206959A1 (en) Orexin receptor antagonists
JP2010229096A (en) Pharmaceutical composition

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680023720.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 187634

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2610400

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008515847

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/015427

Country of ref document: MX

Ref document number: 5617/CHENP/2007

Country of ref document: IN

Ref document number: 2006255009

Country of ref document: AU

Ref document number: 2006760716

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006255009

Country of ref document: AU

Date of ref document: 20060607

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0611348

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20071206